EP2225267A1 - Polypeptides comprenant une activité de liaison à la glace - Google Patents

Polypeptides comprenant une activité de liaison à la glace

Info

Publication number
EP2225267A1
EP2225267A1 EP08851310A EP08851310A EP2225267A1 EP 2225267 A1 EP2225267 A1 EP 2225267A1 EP 08851310 A EP08851310 A EP 08851310A EP 08851310 A EP08851310 A EP 08851310A EP 2225267 A1 EP2225267 A1 EP 2225267A1
Authority
EP
European Patent Office
Prior art keywords
polypeptide
seq
polypeptides
polypeptide according
frozen
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
EP08851310A
Other languages
German (de)
English (en)
Other versions
EP2225267B1 (fr
Inventor
Hans RAMLØV
Casper Wilkens
Anders LØBNER-OLESEN
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Roskilde Universitet
Original Assignee
Roskilde Universitet
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Roskilde Universitet filed Critical Roskilde Universitet
Priority to DK12192989.7T priority Critical patent/DK2602263T3/da
Priority to EP12192989.7A priority patent/EP2602263B1/fr
Publication of EP2225267A1 publication Critical patent/EP2225267A1/fr
Application granted granted Critical
Publication of EP2225267B1 publication Critical patent/EP2225267B1/fr
Not-in-force legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • C07K14/435Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans
    • C07K14/43504Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates
    • C07K14/43563Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof from animals; from humans from invertebrates from insects
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N1/00Preservation of bodies of humans or animals, or parts thereof
    • A01N1/02Preservation of living parts
    • A01N1/0205Chemical aspects
    • A01N1/021Preservation or perfusion media, liquids, solids or gases used in the preservation of cells, tissue, organs or bodily fluids
    • A01N1/0221Freeze-process protecting agents, i.e. substances protecting cells from effects of the physical process, e.g. cryoprotectants, osmolarity regulators like oncotic agents
    • AHUMAN NECESSITIES
    • A01AGRICULTURE; FORESTRY; ANIMAL HUSBANDRY; HUNTING; TRAPPING; FISHING
    • A01NPRESERVATION OF BODIES OF HUMANS OR ANIMALS OR PLANTS OR PARTS THEREOF; BIOCIDES, e.g. AS DISINFECTANTS, AS PESTICIDES OR AS HERBICIDES; PEST REPELLANTS OR ATTRACTANTS; PLANT GROWTH REGULATORS
    • A01N3/00Preservation of plants or parts thereof, e.g. inhibiting evaporation, improvement of the appearance of leaves or protection against physical influences such as UV radiation using chemical compositions; Grafting wax
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23BPRESERVING, e.g. BY CANNING, MEAT, FISH, EGGS, FRUIT, VEGETABLES, EDIBLE SEEDS; CHEMICAL RIPENING OF FRUIT OR VEGETABLES; THE PRESERVED, RIPENED, OR CANNED PRODUCTS
    • A23B7/00Preservation or chemical ripening of fruit or vegetables
    • A23B7/04Freezing; Subsequent thawing; Cooling
    • A23B7/05Freezing; Subsequent thawing; Cooling with addition of chemicals or treatment with chemicals other than cryogenics, before or during cooling, e.g. in the form of an ice coating or frozen block
    • A23B7/055Freezing; Subsequent thawing; Cooling with addition of chemicals or treatment with chemicals other than cryogenics, before or during cooling, e.g. in the form of an ice coating or frozen block with direct contact between the food and the chemical, e.g. liquid nitrogen, at cryogenic temperature
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23BPRESERVING, e.g. BY CANNING, MEAT, FISH, EGGS, FRUIT, VEGETABLES, EDIBLE SEEDS; CHEMICAL RIPENING OF FRUIT OR VEGETABLES; THE PRESERVED, RIPENED, OR CANNED PRODUCTS
    • A23B7/00Preservation or chemical ripening of fruit or vegetables
    • A23B7/14Preserving or ripening with chemicals not covered by groups A23B7/08 or A23B7/10
    • A23B7/153Preserving or ripening with chemicals not covered by groups A23B7/08 or A23B7/10 in the form of liquids or solids
    • A23B7/154Organic compounds; Microorganisms; Enzymes
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23GCOCOA; COCOA PRODUCTS, e.g. CHOCOLATE; SUBSTITUTES FOR COCOA OR COCOA PRODUCTS; CONFECTIONERY; CHEWING GUM; ICE-CREAM; PREPARATION THEREOF
    • A23G9/00Frozen sweets, e.g. ice confectionery, ice-cream; Mixtures therefor
    • A23G9/32Frozen sweets, e.g. ice confectionery, ice-cream; Mixtures therefor characterised by the composition containing organic or inorganic compounds
    • A23G9/38Frozen sweets, e.g. ice confectionery, ice-cream; Mixtures therefor characterised by the composition containing organic or inorganic compounds containing peptides or proteins
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L29/00Foods or foodstuffs containing additives; Preparation or treatment thereof
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L3/00Preservation of foods or foodstuffs, in general, e.g. pasteurising, sterilising, specially adapted for foods or foodstuffs
    • A23L3/34Preservation of foods or foodstuffs, in general, e.g. pasteurising, sterilising, specially adapted for foods or foodstuffs by treatment with chemicals
    • A23L3/3454Preservation of foods or foodstuffs, in general, e.g. pasteurising, sterilising, specially adapted for foods or foodstuffs by treatment with chemicals in the form of liquids or solids
    • A23L3/3463Organic compounds; Microorganisms; Enzymes
    • A23L3/3526Organic compounds containing nitrogen
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23LFOODS, FOODSTUFFS, OR NON-ALCOHOLIC BEVERAGES, NOT COVERED BY SUBCLASSES A21D OR A23B-A23J; THEIR PREPARATION OR TREATMENT, e.g. COOKING, MODIFICATION OF NUTRITIVE QUALITIES, PHYSICAL TREATMENT; PRESERVATION OF FOODS OR FOODSTUFFS, IN GENERAL
    • A23L3/00Preservation of foods or foodstuffs, in general, e.g. pasteurising, sterilising, specially adapted for foods or foodstuffs
    • A23L3/36Freezing; Subsequent thawing; Cooling
    • A23L3/37Freezing; Subsequent thawing; Cooling with addition of or treatment with chemicals
    • A23L3/375Freezing; Subsequent thawing; Cooling with addition of or treatment with chemicals with direct contact between the food and the chemical, e.g. liquid nitrogen, at cryogenic temperature
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K8/00Cosmetics or similar toiletry preparations
    • A61K8/18Cosmetics or similar toiletry preparations characterised by the composition
    • A61K8/30Cosmetics or similar toiletry preparations characterised by the composition containing organic compounds
    • A61K8/64Proteins; Peptides; Derivatives or degradation products thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61QSPECIFIC USE OF COSMETICS OR SIMILAR TOILETRY PREPARATIONS
    • A61Q19/00Preparations for care of the skin
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K14/00Peptides having more than 20 amino acids; Gastrins; Somatostatins; Melanotropins; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/63Introduction of foreign genetic material using vectors; Vectors; Use of hosts therefor; Regulation of expression
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N9/00Enzymes; Proenzymes; Compositions thereof; Processes for preparing, activating, inhibiting, separating or purifying enzymes
    • C12N9/10Transferases (2.)
    • C12N9/1085Transferases (2.) transferring alkyl or aryl groups other than methyl groups (2.5)
    • C12N9/1088Glutathione transferase (2.5.1.18)
    • FMECHANICAL ENGINEERING; LIGHTING; HEATING; WEAPONS; BLASTING
    • F16ENGINEERING ELEMENTS AND UNITS; GENERAL MEASURES FOR PRODUCING AND MAINTAINING EFFECTIVE FUNCTIONING OF MACHINES OR INSTALLATIONS; THERMAL INSULATION IN GENERAL
    • F16LPIPES; JOINTS OR FITTINGS FOR PIPES; SUPPORTS FOR PIPES, CABLES OR PROTECTIVE TUBING; MEANS FOR THERMAL INSULATION IN GENERAL
    • F16L58/00Protection of pipes or pipe fittings against corrosion or incrustation
    • AHUMAN NECESSITIES
    • A23FOODS OR FOODSTUFFS; TREATMENT THEREOF, NOT COVERED BY OTHER CLASSES
    • A23VINDEXING SCHEME RELATING TO FOODS, FOODSTUFFS OR NON-ALCOHOLIC BEVERAGES AND LACTIC OR PROPIONIC ACID BACTERIA USED IN FOODSTUFFS OR FOOD PREPARATION
    • A23V2002/00Food compositions, function of food ingredients or processes for food or foodstuffs
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K2319/00Fusion polypeptide
    • C07K2319/20Fusion polypeptide containing a tag with affinity for a non-protein ligand
    • C07K2319/23Fusion polypeptide containing a tag with affinity for a non-protein ligand containing a GST-tag
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12YENZYMES
    • C12Y205/00Transferases transferring alkyl or aryl groups, other than methyl groups (2.5)
    • C12Y205/01Transferases transferring alkyl or aryl groups, other than methyl groups (2.5) transferring alkyl or aryl groups, other than methyl groups (2.5.1)
    • C12Y205/01018Glutathione transferase (2.5.1.18)

Definitions

  • Polypeptides comprising an ice-binding activity
  • the present invention relates to novel polypeptides comprising an ice-binding capability resulting in an ice crystal formation and/or growth reducing or inhibiting activity. Methods for producing and using such polypeptides are also disclosed.
  • the body temperature of marine teleosts living in polar waters is in temperature equilibrium with the surrounding sea, and is thus at a temperature of approximately -1 .8 Q C during winter or year round in for example Antarctic waters.
  • the blood of marine teleosts is hypoosmotic to the seawater and its melting point is predicted to be approximately -0.7 Q C.
  • the polar teleosts are thus supercooled and lethal freezing would be expected in the absence of an adaptation to such harsh climatic conditions.
  • Psychrophilic (cold-loving) organisms have successfully adapted to all the permanently cold regions on earth: They can be found in the deep sea, on freezing mountain tops and in the Polar regions even at temperatures as low as -60 Q C. Despite the lethal conditions, these organisms have overcome key barriers inherent to permanently cold environments. These barriers include: reduced enzyme activity, decreased membrane fluidity, altered transport of nutrients and waste products, decreased rates of transcription, translation and cell division, polypeptide cold- denaturation, inappropriate polypeptide folding and intracellular ice formation.
  • Anti-freeze polypeptides (AFP - in some publications also known as thermal hysteresis polypeptides, THP, or ice structuring polypeptides, ISP) lower the freezing point of a solution substantially while the predicted melting point is only moderately depressed. This means that whereas the freezing point is lowered dramatically, the melting point of the solution is predicted by the colligative melting point depression. This is true for solutions where ice is present- the question as to whether anti-freeze polypeptides can lower the supercooling point of an ice-free solution is largely unsolved.
  • THP thermal hysteresis polypeptides
  • ISP ice structuring polypeptides
  • the displacement of the freezing temperature is limited and rapid ice growth will take place at a sufficiently low temperature.
  • the separation of the melting and freezing temperature is usually referred to as thermal hysteresis (TH) (Knight et al. 1991 , Raymond and DeVries 1977, Wilson 1993), and the temperature of ice growth is referred to as the hysteresis freezing point.
  • the difference between the melting point and the hysteresis freezing point is called the hystersis or the anti-freeze activity.
  • a second functionality of the AFPs is in the frozen state, where they show ice recrystallization inhibition (Rl). The AFPs inhibit the formation of large crystals at the expense of small crystals at temperatures above the temperature of recrystrallisation. (Knight et 1.1984, 1995, Knight and Duman 1986, Raml ⁇ v et al. 1996).
  • AFPs seem all to be amphiphilic. This means that they have one part which is more hydrophobic than the rest of the molecule. Hitherto the explanation for their activity is that their hydrophilic side binds to the ice crystal. However, this view has during the last decade been challenged as when looking at ice/water one can with good reason ask which is per definition most hydrophilic- ice or water.
  • Various evidence for the binding of the AFPs to the ice via their hydrophobic side/domains is emerging, but as the exact mechanism for the binding is not known all evidence so far has been circumstantial.
  • the hysteresis freezing point is thus the temperature where it again becomes energetically favourable for the water molecules to bind to the ice and ice growth continues explosively (Knight et al. 1991 , Raymond and DeVries 1977, Wilson 1993). In most fish anti-freeze solutions, spicular ice growth is seen at the hysteresis freezing point. It is assumed that this is due to the fact that the fish anti-freeze polypeptides bind to the prismplanes on the ice crystals but not to the basal planes.
  • the thermal hysteresis is thus dependent on at least 2 parameters: 1 ) the type of anti-freeze polypeptide (dependent on organism, isoform) and 2) the concentration, albeit that the concentration dependency shows saturation (DeVries 1983, Kao et al. 1985, Schrag et al. 1982).
  • AFP is very well characterized is the AFP from the beetle Tenebrio molitor. This polypeptide is found in a least 9 isoforms which are all very much alike, but with lengths from 84, 96 to 120 amino acids (Liou et al. 1999). In all the 9 known isoforms a repetitive sequence of 12 amino acids is found: T/ A-C- T-X-S-X-X-C-X-X-A-X. This sequence is repeated 6 to 9 times in these AFPs. TmAFP is a right handed beta-helix structure of repetitive amino acid sequences.
  • a highly regular array of threonine residues on the flat beta sheet is thought to interact with water/ice, as the distances between the threonine residues is predicted to fit exactly with the oxygen atom positions in the water molecules of the ice lattice structure.
  • This AFP has an extremely regular structure and a predicted high structural stability provided by a high number for cysteine-cysteine sulphur bridges; every sixth amino acid is a cysteine. From D. canadensis 13 isoforms with varying length and weight (7.3 - 12.3 kDa) have been isolated (Andorfer and Duman 2000, Duman et al. 2002). In these AFPs the repeating sequence is the same as is found in T.
  • the amino acid sequences of the AFPs from C. fumiferana are not homologous to the two other known AFP amino acid sequences from insects. However, a sequence T-X-T is found at every 15th amino acid but only the last threonine is conserved; the first Threonine are in many cases substituted with valine (V), arginine (R) or isoleucine (I).
  • the AFPs from C. fumiferana also contain fewer cysteines than the other two insect AFPs (Doucet et al. 2000). Howvever, all cysteines participate in disulfide bindings (Gauthier et al. 1998). Apparently the AFPs from C.
  • fumiferana contains a hydrophobic core and the polypeptides are stabilized by a network of hydrogen bonds and the disulfide bridges, which all together stabilises the structure (Graether et al. 2000, 2003, Leinala et al 2002).
  • the present invention is directed in one aspect to anti-freeze polypeptides and fragments thereof, including anti-freeze polypeptides produced by certain Cerambycid bark beetles and comprising a plurality of ice-binding sites. Methods for making and using such polypeptides, as disclosed herein below in more detail, are also within the scope of the present invention.
  • Rhagium inquisitor and Rhagium mordax both in their adult and larval stages, can be subjected to temperatures as low as -30 Q C and still overwinter under bark on tree stumps in northern Scandinavia. Both of these species belong to the group of cold tolerant animals called freeze avoiders, which means that they survive the extremely low temperatures without ice formation in their tissues. They can be regarded to adopt a supercooled (or undercooled) conditions for long time periods.
  • the supercooled state is potentially lethal to the Cerambycid bark beetles as ice formation in a metastable, supercooled liquid can in principle occur due to random nucleation or as a consequence of inoculation of ice from the surroundings.
  • the Cerambycid bark beetles cannot survive such an ice formation.
  • both R inquisitor and R mordax have adapted to the climatic conditions and they are able to survive at low temperatures for extended time periods.
  • Larvae of R mordax are capable of surviving almost as cold temperatures as R inquisitor- in spite of having a very limited accumulation of cryoprotective compounds. According to one presently preferred hypothesis, the ability of Rhagium mordax to withstand low temperatures can almost exclusively be ascribed to the synthesis and accumulation of one or more anti-freeze polypeptides (AFP's) prior to the onset of winter.
  • AFP's anti-freeze polypeptides
  • Anti-freeze polypeptides according to the present invention are surprisingly found to have a significantly lower number of cysteine residues than other insect AFPs presently known. This feature, together with the fact that the polypeptides according to the present invention have fewer repeated sequences than many state-of-the-art insect anti-freeze polypeptides, make them better candidates for expression in heterologous host organisms.
  • the anti-freeze polypeptides according to the present invention preferably have less than 4 cysteine residues, such as less than 3 cysteine residues, for example 2 cysteine residues.
  • the anti-freeze polypeptides according to the present invention have a variety of utilities and industrial applications as will be clear from the below disclosure.
  • the polypeptides, or genes encoding the polypeptides can be used in various ways to suppress ice crystal growth.
  • the polypeptides may be introduced directly, or they may be introduced as a gene which is expressed in a host cell under the control of a suitable expression signal to produce the polypeptide(s).
  • Suitable concentrations of anti-freeze polypeptides will vary depending on the use, but will typically be in the range of from about one part per billion to about one part per thousand (i.e., from about 1 ⁇ g/l to about 1 mg/l).
  • the polypeptides are introduced into edible products, or brought into contact with edible products, so as to reduce or inhibit ice crystal growth and/or formation e.g. during production and/or storage of the edible products in their frozen condition.
  • the polypeptides according to the present invention provides ice crystallisation that are markedly different than crystals obtained in the presence of other known anti-freeze polypeptides.
  • crystals with a small spheric structure are obtained while known anti-freeze polypeptides such as e.g. anti-freeze protein type III HPLC 12 mentioned in US 6,914,043 or the ice crystal growth inhibiting agent as mentioned in US 6,312,733 provides ice crystals with a spicular structure.
  • one major advantage of the present invention is that when the polypeptides according to the present invention are incorporated into e.g. ice cream an improved mouth feel is obtained due to the fact that the crystals formed in the ice cream during production and storage will have an essentially small spheric structure compared to the rough spicular crystals obtained when using known anti-freeze polypeptides of type III of herein above.
  • the texture, taste, and useful storage life of frozen edible products, including vegetables will be greatly improved as a result of the action of the polypeptides according to the present invention.
  • frozen vegetables such as e.g. celery, potatoes, asparagus, peas, carrots, beans, broccoli, sweet corn, spinach, and the like
  • the texture, taste and useful storage life of various frozen fruits will be enhanced, including strawberries, blueberries, raspberries, citrus fruits, bananas, grapes, kiwis, peaches, pineapples, plums, cherries, tomatoes and mangoes.
  • the introduction into vegetables, and other edible products can be accomplished e.g. by genetic introduction of appropriate polynucleotides into the target organism.
  • Expression can also be on a tissue specific basis. For example, linkage to ripening genes in fruits may result in expression even after harvesting from the producing plant.
  • polypeptides in one important aspect of the invention, are added to foods which are expected to be or remain frozen until, or even during, consumption - and in particular edible products which are consumed in a frozen or cold state.
  • frozen food products are intended to be consumed in the frozen or cold state, for example, ice cream, frozen yogurt, ice milk, sherbet, popsicles, frozen whipped cream, frozen cream pies, frozen puddings and the like.
  • texture and flavour are adversely affected by the formation of large ice crystals throughout a freeze-thaw cycle that occurs in most home frost-free freezers, or upon sustained storage in the frozen state.
  • This ice crystal growth process may be reduced or even prevented entirely, or at least minimized, by using the anti-freeze polypeptides according to the present invention.
  • the anti-freeze polypeptides according to the present invention may be either incorporated throughout the edible product, and/or they may, i.e. such as alternatively, be applied to the surface of the edible product, where condensation and ice crystal formation is expected to occur most readily.
  • yeasts including bakers yeast, comprising polynucleotides encoding the polypeptides according to the present invention.
  • Methods related to this aspect include methods for transform dough yeast with polynucleotides encoding these polypeptides. Upon incorporation and expression of the polynucleotides into the yeasts, and use of these yeasts e.g. in frozen dough, the dough will naturally leaven upon thawing because the yeast viability will remain high upon thawing.
  • An alternative way of incorporating anti-freeze polypeptides into frozen, fermented edible products is to have the organism responsible for the fermentation process produce the anti-freeze polypeptides while fermenting the food.
  • the present invention also embraces methods for preparing a frozen fermented food product.
  • This method comprises the steps of (a) contacting a food product with a microorganism that is capable of secreting a polypeptide according to the present invention, wherein the microorganism is capable of fermenting the food product to produce the fermented food product, (b) incubating the food product with the microorganism under conditions in which fermentation takes place so that a fermented food product is produced having anti-freeze polypeptides according to the present invention present in an amount effective for inhibiting ice crystal growth and/or formation in the product or on the surface of the product; and (c) freezing the fermented food product at a temperature of preferably below -5°C, so as to produce a frozen, fermented food product.
  • Yet another aspect of the present invention is directed to the introduction of antifreeze polypeptides according to the present invention present into biological cells, or extracts thereof destined for frozen storage.
  • biological cells or extracts thereof destined for frozen storage.
  • bacterial cells, yeast cells, plant cells and animal cells comprising the anti-freeze polypeptides according to the present invention present have an increased cell or tissue viability with minimal or no loss of inherent characteristics due to the freeze-thaw process.
  • Subcellular samples or cellular extracts may have similar sensitivities to freezing, especially on prolonged storage.
  • Typical examples will be in vitro polypeptide translation systems, enzyme preparations, and particularly samples which contain sensitive membrane components, such as chloroplast or mitochondrial membrane preparations.
  • samples containing organelles may display increased resistance to freezing damage upon addition of the anti-freeze polypeptides according to the present invention present.
  • Soft animal tissues will exhibit less damage upon freezing in the presence of the subject polypeptides, and addition of the polypeptides according to the present invention present will be useful in situations, when cellular integrity upon freezing and subsequent thawing is important or desired, such as for tissue culture deposits.
  • samples destined for frozen storage such as for cell or tissue depositories, might routinely have the polypeptides according to the present added to them.
  • the biological cell types often stored are genetic variants of bacteria, fungi (including yeast), and, particularly, higher eucaryote cells (such as hybridoma strains and tissue culture cell lines).
  • the present invention in other aspects are directed to applications which are not specific to the food area.
  • One non-food application of the polypeptides according to the present invention present is the protection of crops and plants from climatic freezing conditions.
  • the anti-freeze polypeptides according to the present invention present may be either internally incorporated into the cytoplasm by expression of an introduced gene, or the polypeptides may be externally applied to the plants - e.g. by spraying or otherwise. External application may thus be achieved either by direct application of the polypeptides to the plant, or by the external deposit onto the plant of an organism which secretes the polypeptide.
  • Another embodiment is the introduction of an anti-freeze polypeptide into a liquid surrounding an organ, tissue or other biological sample.
  • the anti-freeze polypeptide according to the present invention present should allow short- or long-term storage at a subfreezing temperature, thereby minimizing inherent metabolism or degradation, but with substantially diminished cellular damage from ice crystal growth.
  • Other medically important temperature sensitive biological samples are blood and blood products, therapeutic agents, polypeptide drugs, bioassay reagents and vaccines.
  • the present invention also provides a cosmetic or dermatological preparation which comprises the polypeptides according to the present invention.
  • polypeptides according to the present invention renders possible an effective treatment, but also a prophylaxis of structural and cellular damage in the skin due to cold, which damage with distinct climate- and weather-induced drops in temperature cause changes in the cell physiology in the cell and in the extracellular space through loss of the temperature optima of cellular enzymes, skin damage, skin redness and tight feeling of the skin and increased sensory sensitivities, induced, e.g., by cold, wind and/or UV light, temperature-sensitive skin, negative changes in the skin, the lips and the mucous membranes in the nose and mouth area and the integumentary appendage caused by environmental stress (caused by temperature changes and UV light, smoking, smog, reactive oxygen species, free radicals).
  • compositions and uses based on the mixture of anti-freeze polypeptides according to the present invention with state-of- the-art stabilizers, emulsifiers and surfactants well known to those skilled in the art and other additives. These compounds may be present to inhibit decay, inhibit oxidation, prevent discoloration, inhibit microbial growth, stabilize emulsions and so forth.
  • the present invention is directed in one aspect to polypeptides capable of inhibiting and/or reducing ice crystal formation associated with the freezing or supercooling of an object or substance, including an edible product.
  • Supercooling conditions are conditions allowing the cooling of a substance below the temperature at which a change of state would ordinarily take place (i.e. from a water phase to ice) without such a change of state occurring. Accordingly, the cooling of a liquid below its freezing point without freezing taking place constitutes supercooling and results in a metastable state.
  • polypeptides according to the present invention will interchangeably be denoted anti-freeze polypeptides and, for short, polypeptides, throughout the present description.
  • polypeptide selected from the group consisting of SEQ ID NO:1 to SEQ ID NO:8.
  • the annotation SEQ ID NO:1 to SEQ ID NO:8, and other similar annotations indicating a starting number and an end number of a range of sequence identity numbers, shall, when used herein, denote each and every one of said sequence identity numbers, such as, in the above cited example, the sequence identity numbers SEQ ID NO:1 , SEQ ID NO:2, SEQ ID NO:3, SEQ ID NO:4, SEQ ID NO:5, SEQ ID NO:6, SEQ ID NO:7 and SEQ ID NO:8, unless otherwise noted.
  • Polypeptides comprising or consisting of any of SEQ ID NO:1 to SEQ ID NO:8, fragments thereof having anti-freeze activity, and variants thereof being at least about 75% identical to any of SEQ ID NO:1 to SEQ ID NO:8, or a fragment thereof, also fall within the scope of the present invention.
  • a fragment of the polypeptide comprising or consisting of any of SEQ ID NO:1 to SEQ ID NO:8 having at least 20 amino acids and an ice binding and anti-freeze activity are also disclosed.
  • the fragment preferably has less than 200 amino acids, such as preferably less than 150 amino acids, for example preferably less than 100 amino acids, such as 80 amino acids, for example 60 amino acids.
  • the invention is also directed to polypeptides comprising one or more "ice binding sites” (IBSs) according to the present invention, such as polypeptides comprising one or more of any of SEQ ID NO:9 to SEQ ID NO: 72 - a total of 64 specific "ice binding sites", IBSs, as indicated in Fig 3.
  • IBSs ice binding sites
  • a polypeptide comprising 4 or more sequences, such as 5 or more sequences, for example 6 or more sequences, such as 7 or all of the sequences selected from the group consisting of SEQ ID NO: 9 to SEQ ID NO:16.
  • a polypeptide comprising 4 or more sequences, such as 5 or more sequences, for example 6 or more sequences, such as 7 or all of the sequences selected from the group consisting of SEQ ID NO: 33 to SEQ ID NO:40.
  • a polypeptide comprising 4 or more sequences, such as 5 or more sequences, for example 6 or more sequences, such as 7 or all of the sequences selected from the group consisting of SEQ ID NO: 41 to SEQ ID NO:48.
  • a polypeptide comprising 4 or more sequences, such as 5 or more sequences, for example 6 or more sequences, such as 7 or all of the sequences selected from the group consisting of SEQ ID NO: 49 to SEQ ID NO:56.
  • a polypeptide comprising 4 or more sequences, such as 5 or more sequences, for example 6 or more sequences, such as 7 or all of the sequences selected from the group consisting of SEQ ID NO: 57 to SEQ ID NO:64.
  • a polypeptide comprising 4 or more sequences, such as 5 or more sequences, for example 6 or more sequences, such as 7 or all of the sequences selected from the group consisting of SEQ ID NO: 65 to SEQ ID NO:72.
  • polypeptides comprising one or more of the sequences SEQ ID NO: 73 to 80, such as 4 or more sequences, such as 5 or more sequences, for example 6 or more sequences, such as 7 or all of the sequences of SEQ ID NO: 73 to 80 - in the form of conserved "ice binding domains", IBDs.
  • SEQ ID NO: 81 to 89 relate to further conserved domains which may also be present, singly, or in any combination, in the polypeptides according to the present invention.
  • the present invention is also directed to SEQ ID NO: 90, in the form of a general ice binding domain, general IBD, which can be present a number of times, such as 2 times, 3 times, 4 times, 5 times, 6 times, 7 times, 8 times, more than 8 times and preferably less than 30 times in the polypeptides according to the present invention.
  • Identical sequences can be present a plurality of times, or variants of SEQ ID NO:90 can be present a plurality of times, as disclosed herein below in the section citing items of the present invention.
  • Polypeptides comprising pair-wise combinations of ice binding domains are listed in Table 1 herein below. A total of 64 different combinations are listed - in the form of each and every pair-wise and independent combination of any of the 8 ice binding domains (IBDs) disclosed herein as SEQ ID NO: 73 to SEQ ID NO:80.
  • IBDs ice binding domains
  • the present invention is also directed to a polypeptide comprising 4 or more sequences, such as 5 or more sequences, for example 6 or more sequences, such as 7 or all of the sequences selected from the group consisting of SEQ ID NO: 73 to SEQ ID NO:80
  • polypeptide comprising 4 or more copies, such as 5 or more copies, for example 6 or more copies, such as 7 or more copies, for example 8 or more copies, and preferably less than 20 copies, of the sequence SEQ ID NO: 90.
  • the present invention in one aspect is directed to a polypeptide comprising a combination of different ice binding domains (IBDs), said combination comprising as least 1 combination, such as at least 2 combinations, of any of the combinations of 2 IBDs selected from the group of 64 pair-wise ice binding domain sequence combinations presented in Table 1 , herein immediately below, said polypeptide preferably having less than 20 general ice binding domains, such as less than 15 general ice binding domains, for example less than 12 general ice binding domains, such as less than 10 general ice binding domains defined by SEQ ID NO:90.
  • IBDs ice binding domains
  • IBD I represents SEQ ID NO 73
  • IBD Il represents SEQ ID NO 74
  • IBD III represents SEQ ID NO 75
  • IBD IV represents SEQ ID NO 76
  • IBD V represents SEQ ID NO 77
  • IBD Vl represents SEQ ID NO 78
  • IBD VII represents SEQ ID NO 79
  • IBD VIII represents SEQ ID NO 80.
  • each of the separate letter/number combinations presented in the matrix denoted Table 1 herein above is cross-combined with each other to produce the combinations listed herein below in Table 2.
  • the combination A1 A1 originates from the combination of two IBD I domains with another two IBD I domains whereas the combination B3A6 originates from the combination of one IBD Il and one IBD III with one IBD I and one IBD Vl.
  • the resulting polypeptide thus comprises the following polypeptide ice binding domains: IBD I, IBD II, IBD III and IBD Vl.
  • the present invention thus further provides a polypeptide comprising a combination of different ice binding domains (IBDs), said combination comprising as least 1 combination, such as at least 2 combinations, for example 3 combinations of any of the combination of 4 IBDs selected from the group of 4096 combinations of any 4 ice-binding domain sequences presented in Table 2, herein immediately below, said polypeptide preferably having less than 20 general ice binding domains, such as less than 15 general ice binding domains, for example less than 12 general ice binding domains, such as less than 10 general ice binding domains defined by SEQ ID NO:90.
  • IBDs ice binding domains
  • the combinations originates from a 64x64 matrix and A1 A1 is the combination of two IBD I domains with another two IBD I domains whereas the combination B3A6 is originated from the combination of one IBD Il and one IBD III domains with one IBD I and one IBD Vl domain.
  • the later thereby consisting of the following polypeptide domains: IBD I, IBD II, IBD III and IBD Vl.
  • E3H7 F1H6 F7H5 G5H4 H3H3 The above-cited polypeptides can further comprise one or more sequences selected from the group consisting of SEQ ID NO: 81 to 89.
  • composition comprising a plurality of identical or different polypeptides as defined herein above and a physiologically acceptable carrier.
  • the composition can be a dried composition and the dried composition can be in freeze dried form or spray dried form.
  • the present invention is directed to a polypeptide having an ice-binding activity and being capable of reducing or inhibiting the growth and/or formation of ice-crystals, wherein the polypeptide comprisies the sequence X1-X2-X3-X4-X5-X6-X7-X8-X9 (SEQ ID NO: 1
  • Xi is selected from the group of amino acid residues consisting of S, A, G and D;
  • X 2 is selected from the group of amino acid residues consisting of A, V, I, T and S;
  • X 3 is selected from the group of amino acid residues consisting of non-bulky amino acid residues
  • X 4 is selected from the group of amino acid residues consisting of S, I, T and V;
  • X 5 is selected from the group of amino acid residues consisting of S, A, I and T;
  • X 6 is selected from the group of amino acid residues consisting of S, T and V;
  • X 7 is selected from the group of amino acid residues consisting of non-bulky amino acid residues
  • X 8 is selected from the group of amino acid residues consisting of S, T and V;
  • X 9 is selected from the group of amino acid residues consisting of S, A and G;
  • the sequence X1-X2-X3-X4-X5-X6-X7-X8-X9 will be referred to herein as a general "ice-binding domain". Whether or not said domain is directly involved in ice- binding or only indirectly involved in ice-binding (i.e. is required in order for the polypeptide to have an ice-binding activity) is immaterial to this definition.
  • the invention further relates to polypeptides comprising a plurality of general "ice- binding domains", such as sequences having a substantial homology/identity to SEQ ID NO:90.
  • a substantial homology to SEQ ID NO:90 shall in this respect encompass any sequence, which differs from the sequence of SEQ ID NO:90 in only one or at the most two of the positions X 1 , X 2 , X 3 , X 4 , X 5 , X 6 ,X?, Xs and X 9 .
  • Plurality as used in this respect shall encompass the intergers 2, 3, 4, 5, 6, 7, 8, 8, 9, such as less than 10, 1 1 , 12, 13, 14, 15, 16, 17, 18, 19, 20, for example less than 25, wherein any of said plurality of general "ice-binding domains" can be identical to or substantially identical to or substantially homologous to SEQ ID NO:90.
  • the invention also relates to modifications and derivatives of the polypeptide according to the present invention comprising one or more copies of SEQ ID NO:90 and optionally further comprising one or more copies of SEQ ID NO:90, or sequences substantially identical or substantially homologous thereto.
  • isolated polypeptide clarifies that the polypeptide according to the present invention is at least essentially free from contaminating cellular components natively associated with the polypeptide.
  • polypeptides according to the present invention can be expressed as fusion polypeptides.
  • Such fusion polypeptides can serve many functions, such as aiding in purification and/or production of the polypeptide in an active conformation.
  • a fusion polypeptide is a polypeptide according to the present invention fused to an affinity tag.
  • the fusion polypeptides can also form part of a complement/anti-complement pair as defined herein, although this term is not limited exclusively to polypeptides. Modifications of the polypeptides, such as splice variants, allelic variants, orthologs and paralogs as defined herein are also within the scope of the present invention. Examples of fusions polypeptides are disclosed herein below in more detail.
  • polypeptides according to the present invention can be labelled with a detactable label, for example a fluorescently detactable label. This can help the practitioner in isolating or identifying the polypeptides according to the present invention.
  • polynucleotides encoding the polypeptides according to the present invention polynucleotide constructs, such as vectors comprising said nucleotides in linear or circular form, host cells transformed with said polynucleotides or vectors comprising said nucleotides, and transgenic organisms comprising said host cells.
  • Polynucleotide is used interchangably with “nucleic acid” and "nucleic acid molecule” unless otherwise indicated.
  • the invention can make use of such a polynucleotide or a complement of such a polynucleotide, for example in the form of a cDNA or an "anti-sense oligonucleotide".
  • the polynucleotide can be a degenerated sequence of individual nucleotides as long as the polynucleotide encodes a polypeptide according to the present invention.
  • the polynucleotide need not comprise all of the individual nucleotides of a native gene as isolated from a natural host organism.
  • Any truncation of such as native gene including sequences which are at least 75% homologous or identical, such as at least 80% homologous or identical, for example at least 85% homologous or identical, such as at least 90% homologous or identical, for example at least 91% homologous or identical, such as at least 92% homologous or identical, for example at least 93% homologous or identical, such as at least 94% homologous or identical, for example at least 95% homologous or identical, such as at least 96% homologous or identical, for example at least 97% homologous or identical, such as at least 98% homologous or identical, for example at least 99% homologous or identical, such as at least 99.5% homologous or identical to the native gene shall be encompassed by the present invention.
  • Any such functional truncation of a native gene including any derivative or modification thereof capable of
  • Expression can be obtained e.g. when a polynucleotide sequence cloned in a cloning vector or expression vector is introduced into a host organism and expressed.
  • the expression is suitably directed by a regulatory sequence typically comprising a promoter which may again comprising elements such as a core promoter and one or more regulatory elements, including an enhancer of expression.
  • the host organism will typically be a recombinant host, i.e. a host not natively harbouring the polynucleotide sequence to be expressed, or not natively comprising the polynucleotide sequence to be expressed operably linked to the native expression signal.
  • a recombinant host i.e. a host not natively harbouring the polynucleotide sequence to be expressed, or not natively comprising the polynucleotide sequence to be expressed operably linked to the native expression signal.
  • a secretory signal sequence When preceded by a secretory signal sequence the polypeptide according to the present invention is destined for secretion - irrespective of whether such a secretion takes place or not.
  • polynucleotide according to the present invention will be referred to as an "isolated" polynucleotide in order to distinguish the polynucleotide from the same or a related sequence in its native environment.
  • heterologous polynucleotide as defined herein signifies a polynucleotide or polynucleotide construct which differs from the native form of the polypeptide according to the present invention.
  • the polynucleotides according to the present invention can be chromosomally integrated or episomal.
  • the invention also provides antibodies, or binding fragments thereof, specific for the polypeptides according to the invention.
  • the antibodies can be produced by any state- of-the-art method and the antibodies can be e.g. a naked antibody, a binding fragment of an antibody, an antibody component, an anti-idiotype antibody, a chimeric antibody and a humanized antibody.
  • Also provided with the scope of the present invention are methods for producing and using both a) polynucleotides according to the present invention, b) polypeptides according to the present invention, and c) antibodies according to the present invention specific for said polypeptides.
  • the antibodies according to the present invention can be used for identifying or partitioning from a population of polypeptides a "target polypeptide” or “target peptide” as defined herein.
  • the "target peptide” can be an antigenic peptide" as defined herein.
  • a solid support comprising the polypeptides and/or the antibodies according to the present invention as well as methods for making and using such a solid support.
  • Figure 1 illustrates the amino acid sequence of a fragment of a R. Inquisitor AFP. Putative ice binding motifs are indicated by black boxes.
  • the forward (F) and reverse (R) primers are given by sequence, and the parts of the polypeptide they are derived from is also indicated.
  • the amino acid sequence has a domain which represents or harbours an ice-binding (IB) motif (consensus A/G-Q/R-G-T-A-T-T-T-T-A-T-G-X-A/G) repeated X times and separated by 1 -8 amino acid residues.
  • IB ice-binding
  • Figure 2 illustrates the full-length cDNA sequences of AFPs 1 -8 from R mordax. Coding sequences for putative signal peptides are underlined.
  • the AFP's corresponding to these cDNAs (AFP 1 -8) mainly differed in their N-terminal part, where AFP4 would appear to be without a signal sequence.
  • the AFPs each contained 8 repeats of an amino acid sequence with the core concensus sequence T-A-T-T-T-A-T. This matches part of the putative IB-motif also observed for R. inquisitor (Fig. 1 ), and it is conceivable that this represents the IB motif of R mordax or at least part of it.
  • FIG. 1 Full length amino acid sequences of AFPs 1 -8 from Rhagium mordax. These sequences are denoted SEQ ID NO:1 to SEQ ID NO:8, respectively. Putative signal sequences are indicated in underscore and putative ice binding motives are indicated in boxes.
  • FIG. 4 Illustration of E. coli cells harbouring full length cDNA's cloned into the expression vector pET26B(+). IPTG can be used to induce synthesis of the pertinent AFP. Initially we have induced synthesis of AFP1 in E. coli. The results demonstrate that AFP1 produced in E. coli confer on the E. coli cells the ability to withstand freezing.
  • FIG. 5 Illustrates an SDS-PAGE gel of wild type (wt) soform RmAFPI and deletion variants ( ⁇ 2- 9, WC (Trp-Cys)).
  • the RmAFP variants were constructed as deletions of the ice binding domains one at a time from the C-terminal of RmAFPI ( ⁇ 2- ⁇ 9) (for example ⁇ 4 indicates that putative ice binding domain 4-9 (see figure 2) has been deleted) and a variant containing a Trp and Cys residue in the C-terminal (WC).
  • pGEX vector system GE Healthcare
  • E. coli strains: Origami, BL21 .
  • the proteins were expressed as a fusion protein to Glutathione-S-transferase (GST) containing a thrombin cleavage site between GST and RmAFP.
  • GST Glutathione-S-transferase
  • Figure 6 Shows the progression of the "ice growth-explosion" which occurs at the hysteresis freezing point in 1 ) a solution of RmAFPI and 2) in serum of the eel pout Zoarces viviparus (a danish fish with type 3 AFP). In both cases a small ice crystal was allowed to anneal in the solution before the temperature was lowered. When decreasing the temperature in the RmAFP solution the initial ice crystal (at the arrow in 1A) does not grow or change shape before the hysteresis freezing point is reached. At the hysteresis freezing point (1 B) the ice crystal bursts and due to the super cooled surroundings the solution freezes.
  • the ice growth pattern at the hysteresis freezing point is cauliflower like (1 B, 1 C). This is in contrast to the events seen in 2) (the Z viviparus serum) where, upon cooling, the ice crystal slowly changes shape and become a bipyramid with a hexagonal base plane (2A). At the hysteresis freezing point the ice growth is spicular (2B) and all ice in the solution is growing as spicules (2C).
  • Figure 7 Shows the molecular weight determination of cloned and purified Rhagium mordax AFP 1 .
  • FIG. 8 Shows dimerization of rRmAFPI .
  • A Under native conditions rRmAFP behaves as a dimer, since when it is passed over a size exclusion column (Superdex 75, 10/30, GE Healthcare) it has shorter retention times than other proteins in the same Mw range. Mw estimation from SEC of rRmAFPI gave 28 KDa when the Superdex 75 was calibrated with bovine serum albumin (68 KDa, BSA), trypsin (25 KDa), and human cystatin C (13 KDa).
  • B Likewise RmAFP behaves as a dimer when run in an SDS PAGE giving an estimated Mw of 28 KDa.
  • Lane 1 Bovine serum albumin (BSA); 2: Trypsin; 3: RNAse A; 4 Cystatin C; 5: Lysozyme; 6 RmAFPI ; 7: Glutathione S transferase (GST).
  • BSA Bovine serum albumin
  • 2 Trypsin
  • 3 RNAse A
  • 5 Lysozyme
  • 6 RmAFPI RmAFPI
  • 7 Glutathione S transferase (GST).
  • Figure 9 Shows a circular dichroism spectroscopy (CD) of a polypeptide according to the invention.
  • Figure 10 Shows the temperature stability of RmAFPI within the temperature range 6 - 100 °C and succeeding 1 1 scans were performed within the temperature range 6 -70 °C.
  • Figure 1 Show the influence of pH on activity and stability of RmAFPI . Definitions
  • Antifreeze proteins lower the freezing temperature of a solution noncolligatively by binding to ice crystals and inhibiting crystal growth, but the proteins alter the melting temperature of the solution only by colligative effects.
  • This thermal hysteresis (the difference between freezing and melting temperatures) is determined by observing the effect of temperature on the growth of a single ice crystal. Melting occurs when faces of the ice crystal become round; freezing occurs when the ice crystal elongates along its c- axis.
  • anti-freeze activity refers to the separation of the melting and freezing temperature. It also refers to the difference between melting point and the freezing point. It further refers to the inhibition of formation of large crystals at the expense of small crystals at temperatures above the temperature of recrystallisation.
  • anti-freeze activity can be used interchangeably with thermal hysteresis.
  • nucleic acid or “nucleic acid molecule” refers to polynucleotides, such as deoxyribonucleic acid (DNA) or ribonucleic acid (RNA), oligonucleotides, fragments generated by the polymerase chain reaction (PCR), and fragments generated by any of ligation, scission, endonuclease action, and exonuclease action, polynucleotide molecules can be composed of monomers that are naturally-occurring nucleotides (such as DNA and RNA), or analogs of naturally-occurring nucleotides (e.g., (alpha- enantiomeric forms of naturally-occurring nucleotides), or a combination of both.
  • DNA deoxyribonucleic acid
  • RNA ribonucleic acid
  • PCR polymerase chain reaction
  • Modified nucleotides can have alterations in sugar moieties and/or in pyrimidine or purine base moieties.
  • Sugar modifications include, for example, replacement of one or more hydroxyl groups with halogens, alkyl groups, amines, and azido groups, or sugars can be functionalized as ethers or esters.
  • the entire sugar moiety can be replaced with sterically and electronically similar structures, such as aza-sugars and carbocyclic sugar analogs.
  • Examples of modifications in a base moiety include alkylated purines and pyrimidines, acylated purines or pyrimidines, or other well-known heterocyclic substitutes, polynucleotide monomers can be linked by phosphodiester bonds or analogs of such linkages. Analogs of phosphodiester linkages include phosphorothioate, phosphorodithioate, phosphoroselenoate, phosphorodiselenoate, phosphoroanilothioate, phosphoranilidate, phosphoramidate, and the like.
  • nucleic acid molecule also includes so-called “peptide polynucleotides,” which comprise naturally-occurring or modified polynucleotide bases attached to a polyamide backbone, polynucleotides can be either single stranded or double stranded.
  • complement of a polynucleotide molecule refers to a polynucleotide molecule having a complementary nucleotide sequence and reverse orientation as compared to a reference nucleotide sequence.
  • sequence 5' ATGCACGGG 3' is complementary to 5' CCCGTGCAT 3'.
  • degenerate nucleotide sequence denotes a sequence of nucleotides that includes one or more degenerate codons as compared to a reference polynucleotide molecule that encodes a polypeptide.
  • Degenerate codons contain different triplets of nucleotides, but encode the same amino acid residue (i.e., GAU and GAC triplets each encode Asp).
  • structural gene refers to a polynucleotide molecule that is transcribed into messenger RNA (mRNA), which is then translated into a sequence of amino acids characteristic of a specific polypeptide.
  • an "isolated polynucleotide molecule” is a polynucleotide molecule that is not integrated in the genomic DNA of an organism.
  • a DNA molecule that encodes a growth factor that has been separated from the genomic DNA of a cell is an isolated DNA molecule.
  • Another example of an isolated polynucleotide molecule is a chemically-synthesized polynucleotide molecule that is not integrated in the genome of an organism.
  • a polynucleotide molecule that has been isolated from a particular species is smaller than the complete DNA molecule of a chromosome from that species.
  • a "nucleic acid molecule construct” is a polynucleotide molecule, either single- or double-stranded, that has been modified through human intervention to contain segments of polynucleotide combined and juxtaposed in an arrangement not existing in nature.
  • Linear DNA denotes non-circular DNA molecules having free 5' and 3' ends.
  • Linear DNA can be prepared from closed circular DNA molecules, such as plasmids, by enzymatic digestion or physical disruption.
  • cDNA complementary DNA
  • cDNA is a single-stranded DNA molecule that is formed from an mRNA template by the enzyme reverse transcriptase. Typically, a primer complementary to portions of mRNA is employed for the initiation of reverse transcription.
  • cDNA to refer to a double- stranded DNA molecule consisting of such a single-stranded DNA molecule and its complementary DNA strand.
  • the term “cDNA” also refers to a clone of a cDNA molecule synthesized from an RNA template.
  • a “promoter” is a nucleotide sequence that directs the transcription of a structural gene.
  • a promoter is located in the 5' non-coding region of a gene, proximal to the transcriptional start site of a structural gene. Sequence elements within promoters that function in the initiation of transcription are often characterized by consensus nucleotide sequences. These promoter elements include RNA polymerase binding sites, TATA sequences, CAAT sequences, differentiation-specific elements (DSEs; McGehee et al., MoI. Endocrinol. 7:551 (1993)), cyclic AMP response elements (CREs), serum response elements (SREs; Treisman, Seminars in Cancer Biol.
  • DSEs differentiation-specific elements
  • CREs cyclic AMP response elements
  • SREs serum response elements
  • GREs glucocorticoid response elements
  • binding sites for other transcription factors such as CRE/ATF (O'Reilly et al., J. Biol. Chem. 267:19938 (1992)), AP2 (Ye et al., J. Biol. Chem. 269:25728 (1994)), SP1 , cAMP response element binding polypeptide (CREB; Loeken, Gene Expr. 3:253 (1993)) and octamer factors (see, in general, Watson et al., eds., Molecular Biology of the Gene, 4th ed. (The Benjamin/Cummings Publishing Company, Inc.
  • a promoter is an inducible promoter, then the rate of transcription increases in response to an inducing agent. In contrast, the rate of transcription is not regulated by an inducing agent if the promoter is a constitutive promoter. Repressible promoters are also known.
  • a “core promoter” contains essential nucleotide sequences for promoter function, including the TATA box and start of transcription. By this definition, a core promoter may or may not have detectable activity in the absence of specific sequences that may enhance the activity or confer tissue specific activity.
  • a “regulatory element” is a nucleotide sequence that modulates the activity of a core promoter.
  • a regulatory element may contain a nucleotide sequence that binds with cellular factors enabling transcription exclusively or preferentially in particular cells, tissues, or organelles. These types of regulatory elements are normally associated with genes that are expressed in a "cell-specific,” “tissue-specific,” or “organelle-specific” manner.
  • An “enhancer” is a type of regulatory element that can increase the efficiency of transcription, regardless of the distance or orientation of the enhancer relative to the start site of transcription.
  • Heterologous DNA refers to a DNA molecule, or a population of DNA molecules, that does not exist naturally within a given host cell.
  • DNA molecules heterologous to a particular host cell may contain DNA derived from the host cell species (i.e., endogenous DNA) so long as that host DNA is combined with non-host DNA (i.e., exogenous DNA).
  • a DNA molecule containing a non-host DNA segment encoding a polypeptide operably linked to a host DNA segment comprising a transcription promoter is considered to be a heterologous DNA molecule.
  • a heterologous DNA molecule can comprise an endogenous gene operably linked with an exogenous promoter.
  • a DNA molecule comprising a gene derived from a wild-type cell is considered to be heterologous DNA if that DNA molecule is introduced into a mutant cell that lacks the wild-type gene.
  • a "polypeptide” is a polymer of amino acid residues preferably joined exclusively by peptide bonds, whether produced naturally or synthetically.
  • a polypeptide produced by expression of a non-host DNA molecule is a "heterologous" peptide or polypeptide.
  • An "amino acid residue” can be a natural or non-natural amino acid residue linked peptide bonds or bonds different from peptide bonds. The amino acid residues can be in D-configuration or L-configuration.
  • a “homopolymer” is a polypeptide which is built up by adding several similar polypeptides to an original polypeptide thereby creating multiple copies of the same polypeptide as one larger polypeptide.
  • a “heteropolymer” is a polypeptide which is built up by adding several different polypeptides to an original polypeptide thereby creating multiple copies of the different polypeptide as one larger polypeptide.
  • a “non-bulky amino acid residue” is preferably a natural amino acid excluding amino acids having either a cyclic (aliphatic or aromatic) side chain, such as e.g. Pro, Phe, Trp, Tyr and His, or a long or branched aliphatic side chain, such as e.g. Arg, Lys, Leu, lie, Met and VaI, or more generally, a bulky amino acid has a side chain having at least 3 carbons, which are linked and form a branched or unbranched side chain.
  • a bulky amino acid has a side chain having at least 3 carbons, which are linked and form a branched or unbranched side chain.
  • Presently preferred examples of "non-bulky amino acids” comprise GIy, Ala and Ser.
  • polypeptide according to the present invention is any polypeptide cited in the claims of the present patent application or the patent granted on the basis of claims of this patent application.
  • a "polynucleotide according to the present invention” or a “nucleic acid according to the present invention” is any polynucleotide encoding a "polypeptide according to the present invention", including any polypeptide cited in the claims of the present patent application or the patent granted on the basis of claims of this patent application.
  • integrated genetic element is a segment of DNA that has been incorporated into a chromosome of a host cell after that element is introduced into the cell through human manipulation.
  • integrated genetic elements are most commonly derived from linearized plasmids that are introduced into the cells by electroporation or other techniques. Integrated genetic elements are passed from the original host cell to its progeny.
  • a "cloning vector” is a polynucleotide molecule, such as a plasmid, cosmid, or bacteriophage, that has the capability of replicating autonomously in a host cell.
  • Cloning vectors typically contain one or a small number of restriction endonuclease recognition sites that allow insertion of a polynucleotide molecule in a determinable fashion without loss of an essential biological function of the vector, as well as nucleotide sequences encoding a marker gene that is suitable for use in the identification and selection of cells transformed with the cloning vector.
  • Marker genes typically include genes that provide tetracycline resistance or ampicillin resistance.
  • an “expression vector” is a polynucleotide molecule encoding a gene that is expressed in a host cell.
  • an expression vector comprises a transcription promoter, a gene, and a transcription terminator. Gene expression is usually placed under the control of a promoter, and such a gene is said to be “operably linked to” the promoter.
  • a regulatory element and a core promoter are operably linked if the regulatory element modulates the activity of the core promoter.
  • a “recombinant host” is a cell that contains a heterologous polynucleotide molecule, such as a cloning vector or expression vector.
  • “Integrative transformants” are recombinant host cells, in which heterologous DNA has become integrated into the genomic DNA of the cells.
  • a “fusion polypeptide” is a hybrid polypeptide expressed by a polynucleotide molecule comprising nucleotide sequences of at least two genes.
  • a fusion polypeptide can comprise at least part of a polypeptide according to the present invention fused with a polypeptide that binds an affinity matrix.
  • Such a fusion polypeptide provides a means to isolate large quantities of a polypeptide according to the present invention using affinity chromatography.
  • secretory signal sequence denotes a DNA sequence that encodes a peptide (a "secretory peptide") that, as a component of a larger polypeptide, directs the larger polypeptide through a secretory pathway of a cell in which it is synthesized.
  • secretory peptide a DNA sequence that encodes a peptide that, as a component of a larger polypeptide, directs the larger polypeptide through a secretory pathway of a cell in which it is synthesized.
  • the larger polypeptide is commonly cleaved to remove the secretory peptide during transit through the secretory pathway.
  • isolated polypeptide is a polypeptide that is essentially free from contaminating cellular components, such as carbohydrate, lipid, or other polypeptideaceous impurities associated with the polypeptide in nature.
  • a preparation of isolated polypeptide contains the polypeptide in a highly purified form, i.e., at least about 80% pure, at least about 90% pure, at least about 95% pure, greater than 95% pure, or greater than 99% pure.
  • polypeptide preparation contains an isolated polypeptide is by the appearance of a single band following sodium dodecyl sulfate (SDS)-polyacrylamide gel electrophoresis of the polypeptide preparation and Coomassie Brilliant Blue staining of the gel.
  • SDS sodium dodecyl sulfate
  • isolated does not exclude the presence of the same polypeptide in alternative physical forms, such as dimers or alternatively glycosylated or derivatized forms.
  • amino-terminal and “carboxyl-terminal” are used herein to denote positions within polypeptides. Where the context allows, these terms are used with reference to a particular sequence or portion of a polypeptide to denote proximity or relative position. For example, a certain sequence positioned carboxyl-terminal to a reference sequence within a polypeptide is located proximal to the carboxyl terminus of the reference sequence, but is not necessarily at the carboxyl terminus of the complete polypeptide.
  • expression refers to the biosynthesis of a gene product.
  • expression involves transcription of the structural gene into mRNA and the translation of mRNA into one or more polypeptides.
  • splice variant is used herein to denote alternative forms of RNA transcribed from a gene. Splice variation arises naturally through use of alternative splicing sites within a transcribed RNA molecule, or less commonly between separately transcribed RNA molecules, and may result in several mRNAs transcribed from the same gene. Splice variants may encode polypeptides having altered amino acid sequence. The term splice variant is also used herein to denote a polypeptide encoded by a splice variant of an mRNA transcribed from a gene.
  • complement/anti-complement pair denotes non-identical moieties that form a non-covalently associated, stable pair under appropriate conditions.
  • biotin and avidin are prototypical members of a complement/anti- complement pair.
  • Other exemplary complement/anti-complement pairs include receptor/ligand pairs, antibody/antigen (or hapten or epitope) pairs, sense/antisense polynucleotide pairs, and the like.
  • the complement/anti-complement pair preferably has a binding affinity of less than 10 9 M "1 .
  • an "anti-idiotype antibody” is an antibody that binds with the variable region domain of an immunoglobulin.
  • an anti-idiotype antibody binds with the variable region of an anti-antibody, and thus, an anti-idiotype antibody mimics an epitope of a polypeptide according to the present invention.
  • an “antibody fragment” is a portion of an antibody such as F(ab') 2 , F(ab) 2 , Fab', Fab, and the like. Regardless of structure, an antibody fragment binds with the same antigen that is recognized by the intact antibody. For example, an anti-(polypeptide according to the present invention) monoclonal antibody fragment binds an epitope of a polypeptide according to the present invention.
  • antibody fragment also includes a synthetic or a genetically engineered polypeptide that binds to a specific antigen, such as polypeptides consisting of the light chain variable region, "Fv” fragments consisting of the variable regions of the heavy and light chains, recombinant single chain polypeptide molecules in which light and heavy variable regions are connected by a peptide linker (“scFv polypeptides”), and minimal recognition units consisting of the amino acid residues that mimic the hypervariable region.
  • scFv polypeptides peptide linker
  • a “chimeric antibody” is a recombinant polypeptide that contains the variable domains and complementary determining regions derived from a rodent antibody, while the remainder of the antibody molecule is derived from a human antibody.
  • Humanized antibodies are recombinant polypeptides in which murine complementarity determining regions of a monoclonal antibody have been transferred from heavy and light variable chains of the murine immunoglobulin into a human variable domain.
  • a “detectable label” is a molecule or atom which can be conjugated to an antibody moiety to produce a molecule useful for diagnosis.
  • detectable labels include chelators, photoactive agents, radioisotopes, fluorescent agents, paramagnetic ions, or other marker moieties.
  • affinity tag is used herein to denote a polypeptide segment that can be attached to a second polypeptide to provide for purification or detection of the second polypeptide or provide sites for attachment of the second polypeptide to a substrate.
  • affinity tag any peptide or polypeptide for which an antibody or other specific binding agent is available can be used as an affinity tag.
  • Affinity tags include a poly-histidine tract, polypeptide A (Nilsson et al., EMBO J. 4:1075 (1985); Nilsson et al., Methods Enzymol.
  • naked antibody is an entire antibody, as opposed to an antibody fragment, which is not conjugated with a therapeutic agent. Naked antibodies include both polyclonal and monoclonal antibodies, as well as certain recombinant antibodies, such as chimeric and humanized antibodies.
  • antibody component includes both an entire antibody and an antibody fragment.
  • a “target polypeptide” or a “target peptide” is an amino acid sequence that comprises at least one epitope, and that is expressed on a target cell, such as a tumor cell, or a cell that carries an infectious agent antigen.
  • T cells recognize peptide epitopes presented by a major histocompatibility complex molecule to a target polypeptide or target peptide and typically lyse the target cell or recruit other immune cells to the site of the target cell, thereby killing the target cell.
  • antigenic peptide is a peptide, which will bind a major histocompatibility complex molecule to form an MHC-peptide complex which is recognized by a T cell, thereby inducing a cytotoxic lymphocyte response upon presentation to the T cell.
  • antigenic peptides are capable of binding to an appropriate major histocompatibility complex molecule and inducing a cytotoxic T cells response, such as cell lysis or specific cytokine release against the target cell which binds or expresses the antigen.
  • the antigenic peptide can be bound in the context of a class I or class Il major histocompatibility complex molecule, on an antigen presenting cell or on a target cell.
  • RNA polymerase Il catalyzes the transcription of a structural gene to produce mRNA.
  • a polynucleotide molecule can be designed to contain an RNA polymerase Il template in which the RNA transcript has a sequence that is complementary to that of a specific mRNA.
  • the RNA transcript is termed an "anti- sense RNA” and a polynucleotide molecule that encodes the anti-sense RNA is termed an "anti-sense gene.”
  • Anti-sense RNA molecules are capable of binding to mRNA molecules, resulting in an inhibition of mRNA translation.
  • an "anti-sense oligonucleotide specific for a polynucletide encoding a polypeptide according to the present invention” is an oligonucleotide having a sequence (a) capable of forming a stable triplex with a portion of a gene encoding a polypeptide according to the present invention, or (b) capable of forming a stable duplex with a portion of an mRNA transcript of such a gene.
  • variant gene refers to polynucleotide molecules that encode a polypeptide having an amino acid sequence that is a modification of a polypeptide according to the present invention. Such variants include naturally-occurring polymorphisms of genes according to the present invention, as well as synthetic genes that contain conservative amino acid substitutions of the amino acid sequence of a polypeptide according to the present invention. Additional variant forms of genes are polynucleotide molecules that contain insertions or deletions of the nucleotide sequences described herein. A variant gene according to the present invention can be identified by determining whether the gene hybridizes with a polynucleotide molecule having the nucleotide sequence of a polypeptide according to the present invention, or its complement, under stringent conditions.
  • variant genes can be identified by sequence comparison. Two amino acid sequences have "100% amino acid sequence identity” if the amino acid residues of the two amino acid sequences are the same when aligned for maximal correspondence. Similarly, two nucleotide sequences have "100% nucleotide sequence identity” if the nucleotide residues of the two nucleotide sequences are the same when aligned for maximal correspondence. Sequence comparisons can be performed using standard software programs such as those included in the LASERGENE bioinformatics computing suite, which is produced by DNASTAR (Madison, Wis.).
  • a variant gene encodes a polypeptide which can be characterized by its ability to bind specifically to an anti-(polypeptide according to the invention) antibody.
  • allelic variant is used herein to denote any of two or more alternative forms of a gene occupying the same chromosomal locus. Allelic variation arises naturally through mutation, and may result in phenotypic polymorphism within populations. Gene mutations can be silent (no change in the encoded polypeptide) or may encode polypeptides having altered amino acid sequence.
  • allelic variant is also used herein to denote a polypeptide encoded by an allelic variant of a gene.
  • ortholog denotes a polypeptide or polypeptide obtained from one species that is the functional counterpart of a polypeptide or polypeptide from a different species. Sequence differences among orthologs are the result of speciation.
  • Parentalogs are distinct but structurally related polypeptides made by an organism. Paralogs are believed to arise through gene duplication. For example, alpha-globin, beta-globin, and myoglobin are paralogs of each other.
  • the present invention provides in one embodiment an isolated polypeptide comprising or consisting of a sequence of amino acid residues selected from the group consisting of SEQ ID NO:1 ; SEQ ID NO:2; SEQ ID NO:3; SEQ ID NO:4; SEQ ID NO:5; SEQ ID NO:6; SEQ ID NO:7 and SEQ ID NO:8, wherein said polypeptide is capable of reducing or inhibiting the formation and/or growth of ice crystals, or a fragment thereof capable of reducing or inhibiting the formation and/or growth of ice crystals, or a sequence which is at least 75% identical to any of said sequences.
  • an isolated polynucleotide comprising a sequence of nucleotides encoding a polypeptide according to the present invention, wherein said polynucleotide can further comprise an expression signal capable of directing the expression, in a suitable host cell, of the sequence of nucleotides encoding a polypeptide according to the present invention.
  • a vector comprising a polynucleotide according to the present invention capable of expressing a polypeptide according to the present invention.
  • An isolated, recombinant cell can comprise the polynucleotide according to the present invention or the vector according to the present invention or the polypeptide according to the present invention.
  • an edible product comprising the polypeptide according to the present invention, wherein the edible product can be frozen, or in the form of a frozen confectionary product, such as ice cream product, or bread.
  • the present invention also pertains to methods for making or using the polypeptides according to the present invention, including, in one embodiment, a method for producing the polypeptide according to the present invention, said method comprising the steps of
  • step ii) providing a host cell suitable for the production of a polypeptide according to the present invention by recombinant expression of the polynucleotide provided in step i),
  • said fermented, edible product comprises the polypeptide according to the present invention
  • a method for reducing or inhibiting ice crystal formation in a frozen, edible product comprising the steps of
  • a method for reducing or inhibiting ice crystal growth in a frozen, edible product comprising the steps of i) providing a frozen edible product, or one or more ingredients required for the production thereof, and
  • a method for structuring ice crystals in a frozen, edible product comprising the steps of
  • a method for modulating the texture or organoleptic qualities of a frozen, edible product comprising the steps of
  • a method for monitoring ice crystal formation during the manufacture or storage of a frozen, edible product comprising the steps of
  • a method for performing an in vitro fertilisation (IVF) treatment in a female individual comprising the steps of removing one or more oocyte(s) from a female individual, optionally together with a biological sample comprising follicular fluid; freezing the one or more oocyte(s), optionally together with the biological sample, in the presence of a polypeptide according to the present invention; fertilising one or more of the removed oocytes in vitro; and implanting one or more of the fertilized oocytes into the female individual.
  • IVF in vitro fertilisation
  • a method for increasing the likelihood or probability of pregnancy in a female individual comprising the steps of removing one or more oocyte(s) from a female individual, optionally together with a biological sample comprising follicular fluid; freezing the one or more oocyte(s), optionally together with the biological sample, in the presence of a polypeptide according to the present invention; fertilising one or more of the removed oocytes in vitro; and implanting one or more fertilized oocytes into the female individual, wherein the freezing of the one or more oocyte(s) in the presence of the polypeptide according to the present invention reduces ice crystal growth and/or formation on the oocyte(s), or in an environment, wherein the oocyte(s) are present, thereby increasing the likelihood or probability of pregnancy.
  • the sample can further comprise granulosa-lutein cells or follicular cells and optionally also other ovarian cells recovered from the ovarian follicles of the female individual.
  • the sample further comprises frozen cells from the environment of an oocyte.
  • the present invention is in a further embodiment directed to a polypeptide having an ice-binding activity and comprising one or more copies of the sequence X 1 -X 2 -X 3 -X 4 -X 5 - X 6 -X 7 -X 8 -X 9 (SEQ ID NO:90), such as, for example, 2, 3, 4, 5, 6, 7, 8, 9 or 10 individually selected copies of the general ice binding domain SEQ ID NO:90, wherein X 1 is selected from the group of amino acid residues consisting of S, A, G and D;
  • X 2 is selected from the group of amino acid residues consisting of A, V, I, T and S;
  • X 3 is selected from the group of amino acid residues consisting of non-bulky amino acid residues
  • X 4 is selected from the group of amino acid residues consisting of S, I, T and V;
  • X 5 is selected from the group of amino acid residues consisting of S, A, I and T;
  • X 6 is selected from the group of amino acid residues consisting of S, T and V;
  • X 7 is selected from the group of amino acid residues consisting of non-bulky amino acid residues;
  • X 8 is selected from the group of amino acid residues consisting of S, T and V;
  • X 9 is selected from the group of amino acid residues consisting of S, A and G; and wherein at least one of the residues X 2 , X 4 , X 6 and X 8 of SEQ ID NO:90 is T or V; and wherein the maximum number of amino acid residues of the polypeptide is less than 1000.
  • the maximum number of amino acid residues of a polypeptide according to the invention is preferably less than 500, such as less than 400, for example less than 300, such as less than 250, for example less than 240, such as less than 230, for example less than 220, such as less than 210, for example less than 200, such as less than 190, for example less than 180, such as less than 150, for example less than 140, such as less than 130, for example less than 120, such as less than 1 10, for example less than 100, such as less than 95, for example less than 90, such as less than 85, for example less than 80, such as less than 75, for example less than 70, such as less than 65, for example less than 60, such as less than 55, for example less than 50, such as less than 45, for example less than 40, such as less than 30, for example less than 20, such as less than 15.
  • 500 such as less than 400, for example less than 300, such as less than 250, for example less than 240, such as less than 230, for example less than 220
  • the minimum number of amino acid residues of the polypeptide according to the invention may be 10 or more, such as 12 or more, for example 14 or more, such as 16 or more, for example 18 or more, such as 20 or more, for example 22 or more, such as 24 or more, for example 26 or more, such as 28 or more, for example 30 or more, such as 32 or more, for example 34 or more, such as 36 or more, for example 38 or more, such as 40 or more, for example 42 or more, such as 44 or more, for example 46 or more, such as 48 or more, for example 50 or more, such as 55 or more, for example 60 or more, such as 65 or more, for example 70 or more, such as 75 or more, for example 80 or more, such as 85 or more, for example 90 or more, such as 95 or more, for example 100 or more, wherein, when any maximum number and minimum number is paired, the maximum number is larger than the minimum number.
  • the polypeptide according to the invention comprises a plurality of general ice-binding domains each comprising the sequence of SEQ ID NO:90, or a variant or derivative or modification thereof, as described herein elsewhere, and preferably having 250 amino acid residues at most.
  • the invention relates to a polypeptide sequence that comprises a second sequence, in the form of a further independently selected copy of SEQ ID NO:90, wherein the further copy of SEQ ID NO:90 does not overlap with the first copy of SEQ ID NO:90.
  • the invention in a another embodiment relates to a polypeptide which further comprises a third copy of SEQ ID NO: 90 (i.e. the polypeptide comprises three independently seleted copies of SEQ ID NO:90), and in a still further embodiment the polypeptide further comprises a fourth copy of SEQ ID NO:90 (i.e. the polypeptide comprises three independently seleted copies of SEQ ID NO:90).
  • the independently selected copies of SEQ ID NO:90 can be identical or different as disclosed herein elsewhere.
  • SEQ ID NO:90 can be present in any order relative to each other, and any two sequences can be separated by at least 2 amino acid residues, such as at least 3 amino acid residues, for example at least 4 amino acid residues, such as at least 5 amino acid residues, for example at least 6 amino acid residues, such as at least 7 amino acid residues, for example at least 8 amino acid residues, such as at least 9 amino acid residues, for example at least 10 amino acid residues, such as at least 1 1 amino acid residues, for example at least 12 amino acid residues, such as at least 13 amino acid residues, for example at least 14 amino acid residues, such as at least 15 amino acid residues, for example at least 16 amino acid residues, such as at least 17 amino acid residues, for example at least 18 amino acid residues, such as at least 19 amino acid residues, for example at least 20 amino acid residues, such as at least 21 amino acid residues, for example at least 22 amino acid residues, such as at least 23 amino acid residues, for example at least 24 amino acid residues
  • the polypeptide according to the invention can be linked to a carrier, such as a solid support or semi-solid support.
  • a carrier such as a solid support or semi-solid support.
  • the polypeptide can be covalently or non-covalently linked to any such carrier, for example a surface of a material desirably displaying the polypeptides according to the invention.
  • the invention further relates to a polypeptide according to the present invention fused to an affinity tag.
  • affinity tags are known from the litterature and can be selected from the group comprising for example: His-tag, polypeptide A tag,
  • Avidin/streptavidin polypeptide G, GluthationeS-tranferase, dihyfrofolate reductase (DHFR), Green fluorescent polypeptide (GFP), polyarginine, polycysteine, c-myc, calmodulin binding polypeptide, influenzavirus hemagglutinin; maltos binding protein (MBP) (HA).
  • the invention also encompasses polypeptides wherein one or more amino acid residues are modified, wherein said one or more modification(s) are preferably selected from the group consisting of in vivo or in vitro chemical derivatization, such as acetylation or carboxylation, glycosylation, such as glycosylation resulting from exposing the polypeptide to enzymes which affect glycosylation, for example mammalian glycosylating or deglycosylating enzymes, phosphorylation, such as modification of amino acid residues which results in phosphorylated amino acid residues, for example phosphotyrosine, phosphoserine and phosphothreonine.
  • the polypeptide according to the invention can comprise one or more amino acids independently selected from the group consisting of naturally occurring L-amino acids, naturally occurring D-amino acids as well as non-naturally occuring, synthetic amino acids.
  • the invention also relates to polypeptides of the invention where blocking groups are introduced in order to protect and/or stabilize the N- and/or C-termini of the polypeptide from undesirable degradation.
  • blocking groups may be selected from the group comprising branched or non-branched alkyl groups and acyl groups, such as formyl and acetly groups, as well substituted froms thereof, such as the acetamidomethyl.
  • the invention further relates to modifications and derivatives of the polypeptide according to the invention, nucleotides encoding said polypeptides, vectors comprising said nucleotides, host cells transformed with said vectors and transgenic organisms comprising said cells.
  • DSM 19401 E. coli strain JM109 containing plasmid pGEM-T-Easy-RmAFP1
  • Escherichia coli ALO3232 DSM 19402 E. coli strain JM109 containing plasmid pGEM-T-Easy-RmAFP2
  • DSM 19403 E. coli strain JM109 containing plasmid pGEM-T-Easy-RmAFP3
  • DSM 19404 E. coli strain JM109 containing plasmid pGEM-T-Easy-RmAFP4
  • DSM 19405 E. coli strain JM109 containing plasmid pGEM-T-Easy-RmAFP5 Escherichia coli ALO3236
  • DSM 19406 E. coli strain JM109 containing plasmid pGEM-T-Easy-RmAFP6
  • Escherichia coli ALO3237 DSM 19407 E. coli strain JM109 containing plasmid pGEM-T-Easy-RmAFP7
  • DSM 19408 E. coli strain JM109 containing plasmid pGEM-T-Easy-RmAFP8
  • AUSTRALIA The applicant hereby gives notice that the furnishing of a sample of a microorganism shall only be effected priot to the grant of a patent, or to the lapsing, refusal or withdrawal of an application, to a person who is a skilled addressee without an interest in the invention (Regulation 3.25(3) of the Autstralian Patents Regulation).
  • CANADA The applicant requests that, until either a Canadian patent has been issued on the basis of the present application or the application has been refused, or is abandoned and no longer subject to reinstatement, or is withdrawn, the Commissioner of Patents only authorizes the furnishing of a sample of the deposited biological material referred to in the application to an independent expert nominated by the Commissioner.
  • CROATIA The applicant hereby requests that, samples shall be, upon request, made available between the publication of the application and the granting of the patent only to an independent expert.
  • DENMARK The applicant hereby requests that, until the present application has been laid open to public inspection (by the Danish Patent Office), or has been finally decided upon by the Danish Patent office without having been laid open to public inspection, the furnishing of a sample of the deposited biological material referred to in the application shall only be effected to an expert in the art.
  • GERMANY The applicant hereby requests that, until the grant of a patent or from 20 years from the date of filing if the application is refused or withdrawn, a sample shall only be issued to an independent expert nominated by the applicant.
  • ICELAND The applicant hereby requests that until a patent has been granted or a final decision taken by the Icelandic Patent Office concerning the present application, which decision has not resulted in a patent, the furnishing of a sample of the deposited biological material referred to in the application shall only be effected to an expert in the art.
  • SINGAPORE The applicant hereby requests that the furnishing of a sample of the deposited biological material referred to in the application shall only be made available to an expert.
  • SWEDEN The applicant hereby requests that, until the present application has been laid open to public inspection (by the Swedish Patent Office), or has been finally decided upon by the Swedish Patent Office without having been laid open to public inspection, the furnishing of a sample of the deposited biological material referred to in the application shall only be effected to an expert in the art.
  • the present invention also provides isolated polypeptides that have a substantially similar sequence identity to the polypeptides according to the present invention, such as any of SEQ ID NO:1 to SEQ ID NO:8, or their orthologs.
  • substantially similar sequence identity is used herein to denote polypeptides having at least 70%, such as at least 72%, for example at least 74%, such as at least 76%, for example at least 78%, such as at least 80%, for example at least 82%, such as at least 84%, for example at least 86%, such as at least 88%, for example at least 90%, such as at least 91%, for example at least 92%, such as at least 93%, for example at least 94%, such as at least 95%, for example at least 96%, such as at least 97%, for example at least 98%, such as at least 99%, or greater than 99% sequence identity to any of the sequences SEQ ID NO:1 to SEQ ID NO:8, or their orthologs.
  • the present invention also contemplates variant polynucleotide molecules that can be identified using two criteria: a) a determination of the identity or similarity between a polypeptide having the amino acid sequence of any of the sequences SEQ ID NO:1 to SEQ ID NO:8, cf above, and b) a hybridization assay carried out under stringent conditions.
  • certain gene variants comprise polynucleotides that remain hybridized with a polynucleotide encoding a polypeptide according to the present invention, such as any of the sequences SEQ ID NO:1 to SEQ ID NO:8, or a complement of such a polynucleotide, following washing under stringent washing conditions, in which the wash stringency is equivalent to 0.5 X to 2 X SSC with 0.1% SDS at 55 °C to 65°C.
  • variant genes can be characterized as polynucleotide molecules that remain hybridized with a polynucleotide encoding a polypeptide according to the present invention, such as any of the sequences SEQ ID NO:1 to SEQ ID NO:8, or a complement of such a polynucleotide, following washing under stringent washing conditions, in which the wash stringency is equivalent to 0.1 X to 0.2 X SSC with 0.1% SDS at 55 °C to 65°C.
  • Percent sequence identity is determined by conventional methods. See, for example, Altschul et al., Bull. Math. Bio. 48:603 (1986), and Henikoff and Henikoff, Proc. Natl. Acad. Sci. USA 89:10915 (1992). Briefly, two amino acid sequences are aligned to optimize the alignment scores using a gap opening penalty of 10, a gap extension penalty of 1 , and the "BLOSUM62" scoring matrix of Henikoff and Henikoff (ibid.). The percent identity is then calculated as: ([Total number of identical matches]/[length of the longer sequence plus the number of gaps introduced into the longer sequence in order to align the two sequences]) x (100).
  • the "FASTA" similarity search algorithm of Pearson and Lipman is a suitable polypeptide alignment method for examining the level of identity shared by an amino acid sequence disclosed herein and the amino acid sequence of a putative or variant.
  • the FASTA algorithm is described by Pearson and Lipman, Proc. Nat'l Acad. Sci. USA 85:2444 (1988), and by Pearson, Meth. Enzymol. 183:63 (1990).
  • the ten regions with the highest density of identities are then rescored by comparing the similarity of all paired amino acids using an amino acid substitution matrix, and the ends of the regions are "trimmed" to include only those residues that contribute to the highest score.
  • the trimmed initial regions are examined to determine whether the regions can be joined to form an approximate alignment with gaps.
  • the highest scoring regions of the two amino acid sequences are aligned using a modification of the Needleman-Wunsch-Sellers algorithm (Needleman and Wunsch, J. MoI. Biol. 48:444 (1970); Sellers, SIAM J. Appl. Math. 26:787 (1974)), which allows for amino acid insertions and deletions.
  • FASTA can also be used to determine the sequence identity of polynucleotide molecules using a ratio as disclosed above.
  • the ktup value can range between one to six, preferably from three to six, and most preferably, three.
  • substitution of amino acid residues in polypeptides according to the present invention is also directed to polypeptides having one or more conservative amino acid substitution(s) and polynucleotides encoding polypeptides having one or more conservative amino acid substitution(s), as compared with the amino acid sequence of any of the sequences SEQ ID NO:1 to SEQ ID NO:8. That is, variants can be obtained that contain one or more amino acid substitutions of any of the sequences SEQ ID NO:1 to SEQ ID NO:8.
  • Variants include sequences wherein an alkyl amino acid is substituted for an alkyl amino acid, wherein an aromatic amino acid is substituted for an aromatic amino acid, wherein a sulfur-containing amino acid is substituted for a sulfur-containing amino acid in, wherein a hydroxy-containing amino acid is substituted for a hydroxy-containing amino acid, wherein an acidic amino acid is substituted for an acidic amino acid, wherien a basic amino acid is substituted for a basic amino acid, or wherein a dibasic monocarboxylic amino acid is substituted for a dibasic monocarboxylic amino acid.
  • a “conservative amino acid substitution” can also be illustrated by a substitution among amino acids within each of the following groups: (1 ) glycine, alanine, valine, leucine, and isoleucine, (2) phenylalanine, tyrosine, and tryptophan, (3) serine and threonine, (4) aspartate and glutamate, (5) glutamine and asparagine, and (6) lysine, arginine and histidine.
  • the BLOSUM62 table is an amino acid substitution matrix derived from about 2,000 local multiple alignments of polypeptide sequence segments, representing highly conserved regions of more than 500 groups of related polypeptides (Henikoff and Henikoff, Proc. Nat'l Acad. Sci. USA 89:10915 (1992)). Accordingly, the BLOSUM62 substitution frequencies can be used to define conservative amino acid substitutions that may be introduced into the amino acid sequences of the present invention.
  • conservative amino acid substitution preferably refers to a substitution represented by a BLOSUM62 value of greater than - 1.
  • an amino acid substitution is conservative if the substitution is characterized by a BLOSUM62 value of 0, 1 , 2, or 3.
  • preferred conservative amino acid substitutions are characterized by a BLOSUM62 value of at least 1 (e.g., 1 , 2 or 3), while more preferred conservative amino acid substitutions are characterized by a BLOSUM62 value of at least 2 (e.g., 2 or 3).
  • polypeptides are characterized by having at least 70%, at least
  • Variants of amino acid sequences can be obtained, for example, by oligonucleotide-directed mutagenesis, linker-scanning mutagenesis, mutagenesis using the polymerase chain reaction, and the like (see Ausubel (1995) at pages 8 10 to 8 22; and McPherson (ed.), Directed Mutagenesis: A Practical Approach (IRL Press 1991 )).
  • the polypeptides according to the present invention can also comprise non-naturally occurring amino acid residues.
  • Non-naturally occurring amino acids include e.g., without limitation, trans-3-methylproline, 2,4-methanoproline, cis-4-hydroxyproline, trans-4-hydroxyproline, N-methylglycine, allo-threonine, methylthreonine, hydroxyethylcysteine, hydroxyethylhomocysteine, nitroglutamnine, homoglutamine, pipecolic acid, thiazolidine carboxylic acid, dehydroproline, 3- and 4-methylproline, 3,3- dimethylproline, tert-leucine, norvaline, 2-azaphenylalanine, 3-azaphenylalanine, A- azaphenylalanine, and 4-fluorophenylalanine.
  • Variants of the disclosed nucleotide and polypeptide sequences according to the present invention can also be generated through DNA shuffling as disclosed by Stemmer, Nature 370:389 (1994), Stemmer, Proc. Nat'l Acad. Sci. USA 91 :10747
  • variant DNA molecules are generated by in vitro homologous recombination by random fragmentation of a parent DNA followed by reassembly using PCR, resulting in randomly introduced point mutations.
  • This technique can be modified by using a family of parent DNA molecules, such as allelic variants or DNA molecules from different species, to introduce additional variability into the process. Selection or screening for the desired activity, followed by additional iterations of mutagenesis and assay provides for rapid "evolution" of sequences by selecting for desirable mutations while simultaneously selecting against detrimental changes.
  • Mutagenesis methods as disclosed herein can be combined with high-throughput, automated screening methods to detect activity of cloned, mutagenized polypeptides in host cells.
  • Mutagenized DNA molecules that encode biologically active polypeptides, or polypeptides that bind specific antibodies can be recovered from the host cells and rapidly sequenced using modern equipment. These methods allow the rapid determination of the importance of individual amino acid residues in a polypeptide of interest, and can be applied to polypeptides of unknown structure.
  • the present invention also includes "functional fragments" of polypeptides and polynucleotide molecules according to the present invention encoding such functional fragments.
  • Routine deletion analyses of polynucleotide molecules can be performed to obtain functional fragments of a polynucleotide molecule that encodes a polypeptide according to the present inventon.
  • DNA molecules encoding any of the sequences SEQ ID NO:1 to SEQ ID NO:8 can be digested with Bal31 nuclease to obtain a series of nested deletions. The fragments are then inserted into expression vectors in proper reading frame, and the expressed polypeptides are isolated and tested for the ability to bind specifically to anti-antibodies.
  • exonuclease digestion is to use oligonucleotide-directed mutagenesis to introduce deletions or stop codons to specify production of a desired fragment.
  • particular fragments of a gene according to the present inventon can be synthesized using the polymerase chain reaction.
  • Fragments of SEQ ID NO:1 to SEQ ID NO:8 preferably comprises one or more of the ice binding sites denoted SEQ ID NO:9 to SEQ ID NO:72, as described herein elsewhere. Chimeric polypeptides and polypeptide fragments are also provided.
  • GSYSCRAVGVDASTVTDVQGTCHAKATGPGAVASGTSVDGSTSTATATGSC originates from the full length sequences SEQ ID NO 1 , SEQ ID NO 2, SEQ NO 7 and SEQID NO 8 and contains the alternative residues C in the C-terminal end and GS in the N-terminal end.
  • the present invention also contemplates functional fragments of a polypeptide according to the present inventon that have amino acid changes, compared with the amino acid sequence of any of the sequences SEQ ID NO:1 to SEQ ID NO:8.
  • a variant polypeptide can be identified on the basis of structure by determining the level of identity with a particular amino acid sequence disclosed herein.
  • An alternative approach to identifying a variant polypeptide on the basis of structure is to determine whether a polynucleotide molecule encoding a potential variant polypeptide can hybridize to a polynucleotide molecule having the nucleotide sequence of any of the sequences SEQ ID NO:1 to SEQ ID NO:8, as discussed above.
  • the present invention also provides polypeptide fragments or peptides comprising an epitope-bearing portion of a polypeptide according to the present inventon as described herein.
  • Such fragments or peptides may comprise an "immunogenic epitope," which is a part of a polypeptide that elicits an antibody response when the entire polypeptide is used as an immunogen.
  • Immunogenic epitope-bearing peptides can be identified using standard methods (see, for example, Geysen et al., Proc. Nat'l Acad. Sci. USA 81 :3998 (1983)).
  • polypeptide fragments or peptides may comprise an "antigenic epitope," which is a region of a polypeptide molecule to which an antibody can specifically bind.
  • Certain epitopes consist of a linear or contiguous stretch of amino acids, and the antigenicity of such an epitope is not disrupted by denaturing agents. It is known in the art that relatively short synthetic peptides that can mimic epitopes of a polypeptide can be used to stimulate the production of antibodies against the polypeptide (see, for example, Sutcliffe et al., Science 219:660 (1983)). Accordingly, antigenic epitope- bearing peptides and polypeptides of the present invention are useful to raise antibodies that bind with the polypeptides described herein.
  • Antigenic epitope-bearing peptides and polypeptides can contain at least four to ten amino acids, such as at least ten to fifteen amino acids, for example about 15 to about 30 amino acids of any of the sequences SEQ ID NO:1 to SEQ ID NO:8.
  • Such epitope- bearing peptides and polypeptides can be produced by fragmenting a polypeptide according to the present inventon, or by chemical peptide synthesis, as described herein.
  • epitopes can be selected by phage display of random peptide libraries (see, for example, Lane and Stephen, Curr. Opin. Immunol. 5:268 (1993), and Cortese et al., Curr. Opin. Biotechnol.
  • the gene encodes a polypeptide that may be characterized by its ability to bind specifically to an antibody capable of specifically binding to any of the sequences SEQ ID NO:1 to SEQ ID NO:8.
  • Fusion polypeptides comprising anti-freeze polypeptides or ice binding sites or ice binding domains according to the invention
  • the present invention also includes anti-freeze fusion polypeptides.
  • Anti-freeze fusion polypeptides of the present invention may be targeted to a particular cellular compartment or to the extracellular space, to a particular cell or to particular cell types.
  • the anti-freeze segments may be targeted to a particular cellular organelle. Not only will the peptide be directed to the organelle, but the anti-freeze function may remain functional even when surrounded by other polypeptide segments.
  • fusion to antibodies or other molecules having cell specificity in binding the resistance to cellular damage upon freezing can be conferred to those cell types. This technique will also find use in organs. Examples of polypeptides to which the polypeptide according to the present invention can be bound are listed below:
  • Fusion polypeptides comprising polypeptides according to the present invention can thus be used to express a polypeptide according to the present invention in a recombinant host, and to isolate expressed polypeptides.
  • One type of fusion polypeptide comprises a peptide that guides a polypeptide according to the present invention from a recombinant host cell.
  • a secretory signal sequence also known as a signal peptide, a leader sequence, prepro sequence or pre sequence
  • suitable expression vector is provided in a suitable expression vector.
  • secretory signal sequence may be derived from a polypeptide according to the present invention
  • a suitable signal sequence may also be derived from another secreted polypeptide or synthesized de novo.
  • the secretory signal sequence is operably linked to a gene encoding sequence according to the present invention such that the two sequences are joined in the correct reading frame and positioned to direct the newly synthesized polypeptide into the secretory pathway of the host cell.
  • Secretory signal sequences are commonly positioned 5' to the nucleotide sequence encoding the polypeptide of interest, although certain secretory signal sequences may be positioned elsewhere in the nucleotide sequence of interest (see, e.g., Welch et al., U.S. Pat. No. 5,037,743; Holland et al., U.S. Pat. No. 5,143,830).
  • yeast signal sequence is preferred for expression in yeast cells.
  • suitable yeast signal sequences are those derived from yeast mating phermone alpha-factor (encoded by the MF-alpha1 gene), invertase (encoded by the SUC2 gene), or acid phosphatase (encoded by the PHO5 gene).
  • a heterologous polypeptide in bacterial cells, it is often desirable to express a heterologous polypeptide as a fusion polypeptide to decrease toxicity, increase stability, and to enhance recovery of the expressed polypeptide.
  • a gene according to the present invention can be expressed as a fusion polypeptide comprising a glutathione S-transferase polypeptide.
  • Glutathione S-transferease fusion polypeptides are typically soluble, and easily purifiable from E. coli lysates on immobilized glutathione columns.
  • a fusion polypeptide according to the present invention comprising a maltose binding polypeptide polypeptide can be isolated with an amylose resin column, while a fusion polypeptide comprising the C-terminal end of a truncated polypeptide A gene can be purified using IgG-Sepharose.
  • Established techniques for expressing a heterologous polypeptide as a fusion polypeptide in a bacterial cell are described, for example, by Williams et al., "Expression of Foreign polypeptides in E. coli Using
  • Peptide tags that are useful for isolating heterologous polypeptides expressed by either prokaryotic or eukaryotic cells include polyHistidine tags (which have an affinity for nickel-chelating resin), c-myc tags, calmodulin binding polypeptide (isolated with calmodulin affinity chromatography), substance P, the RYIRS tag (which binds with anti-RYIRS antibodies), the GIu-GIu tag, and the FLAG tag (which binds with anti- FLAG antibodies). See, for example, Luo et al., Arch. Biochem. Biophys. 329:215 (1996), Morganti et al., Biotechnol. Appl. Biochem. 23:67 (1996), and Zheng et al., Gene 186:55 (1997). polynucleotide molecules encoding such peptide tags are available, for example, from Sigma-Aldrich Corporation (St. Louis, Mo.).
  • fusion polypeptide comprises a polypeptide according to the present invention and an immunoglobulin heavy chain constant region, typically an F c fragment, which contains two constant region domains and a hinge region but lacks the variable region.
  • an immunoglobulin heavy chain constant region typically an F c fragment
  • F c fragment an immunoglobulin heavy chain constant region
  • Chang et al., U.S. Pat. No. 5,723,125 describe a fusion polypeptide comprising a human interferon and a human immunoglobulin Fc fragment.
  • the C-terminal of the interferon is linked to the N-terminal of the Fc fragment by a peptide linker moiety.
  • An example of a peptide linker is a peptide comprising primarily a T cell inert sequence, which is immunologically inert.
  • An exemplary peptide linker has the amino acid sequence: GGSGG SGGGG SGGGG S (SEQ ID NO:91 ).
  • a preferred F c moiety is a human gamma4 chain, which is stable in solution and has little or no complement activating activity.
  • the present invention contemplates a fusion polypeptide that comprises a polypeptide according to the present invention, or a fragment thereof, and a human F c fragment, wherein the C- terminus of the polypeptide according to the present invention, or a fragment thereof, is attached to the N-terminus of the F c fragment via a peptide linker.
  • a fusion polypeptide comprising a polypeptide according to the present invention further comprises an IgG sequence.
  • the polypeptide moiety according to the present invention is covalently joined to the amino terminal end of the IgG sequence, and a signal peptide that is covalently joined to the amino terminal of the polypeptide moiety according to the present invention, wherein the IgG sequence comprises or consists of the following elements in the following order: a hinge region, a CH 2 domain, and a CH 3 domain. Accordingly, the IgG sequence lacks a CHi domain.
  • the polypeptide moiety according to the present invention displays an ice-binding activity.
  • Fusion polypeptides can be prepared by methods known to those skilled in the art by preparing each component of the fusion polypeptide and chemically conjugating them. Alternatively, a polynucleotide encoding both components of the fusion polypeptide in the proper reading frame can be generated using known techniques and expressed by the methods described herein. General methods for enzymatic and chemical cleavage of fusion polypeptides are described, for example, by Ausubel (1995) at pages 16 19 to 16 25.
  • Synthesis of anti-freeze polypeptides according to the present invention may be pursued in two forms, either biological or synthetic.
  • the biological method is by expression of polypeptide coding sequence or gene; the synthetic method is by chemical synthesis of a polypeptide.
  • a preferred synthetic method utilizes solid phase peptide synthesis, such as that developed by Merrifield (J. Am. Chem. So ⁇ , (1963) 85:2149-2156). This method will be particularly useful in testing particular compositions or formulations for anti-freeze activity. For large scale production, the biological expression would typically be preferred.
  • the encoding polynucleotide or gene can be a natural gene with recombinant modifications or a totally synthetic sequence that will be expressed in an appropriate expression system.
  • the methods utilized for insertion of a natural sequence segment into an appropriate vector are well known to persons of ordinary skill in the art, see Maniatis or Wu, et al. (1987) Methods in Enzymology, Vol. 153, Academic Press, New York, N. Y.
  • Synthetic sequences can be synthesized by the phosphoramidite chemistry to make particular sections of the sequence (Beaucage and Carruthers, (1981 ) Tet. Letters,
  • Overlapping segments can be synthesized and then ligated together to produce a larger gene.
  • restriction enzyme cutting sites may be introduced which will provide convenient segments which may be easily linked together or inserted to generate tandem repeats, as will be obvious to one of ordinary skill in the art.
  • Purification of the anti-freeze polypeptides will be by methods known to a person of ordinary skill in the art of polypeptide purification.
  • Standard purification techniques may be from either cell lysates or culture medium if the polypeptides are secreted. Typical methods are column chromatography, ammonium sulfate salt precipitations, antibody affinity column chromatography and others.
  • a preferred method of purification is as described by DeVries et al. (1977) Biochem Biophys. Acta 495:388-392.
  • the anti-freeze polypeptides will be purified to substantial homogeneity, usually at least about 70% to 80% pure, preferably about 90-95% pure, most preferably 99% or more pure.
  • the polypeptides will be substantially free of contaminating, naturally associated fish compounds.
  • polypeptides of the present invention can advantageously be produced in recombinant host cells following conventional techniques.
  • a polynucleotide molecule encoding the polypeptide must be operably linked to regulatory sequences that control transcriptional expression in an expression vector and then, introduced into a host cell.
  • expression vectors can include translational regulatory sequences and a marker gene, which is suitable for selection of cells that carry the expression vector.
  • Expression vectors that are suitable for production of a foreign polypeptide in eukaryotic cells typically contain (1 ) prokaryotic DNA elements coding for a bacterial replication origin and an antibiotic resistance marker to provide for the growth and selection of the expression vector in a bacterial host; (2) eukaryotic DNA elements that control initiation of transcription, such as a promoter; and (3) DNA elements that control the processing of transcripts, such as a transcription termination/polyadenylation sequence.
  • expression vectors can also include nucleotide sequences encoding a secretory sequence that directs the heterologous polypeptide into the secretory pathway of a host cell.
  • an expression vector may comprise a gene according to the present invention and a secretory sequence derived from said gene or another secreted gene.
  • vectors commonly used with bacteria include the pET series (Novagen), pGEX series (Ge Healthcare), pBAD-series (Invitrogen).
  • yeasts are the pPic series for Pichia (Invitrogen), the pKlac system from Kluyveromyces lactis (New England biolabs), S. cereviseae vectors (Patel, O., Fearnley, R., and Macreadie, I.. 3002. Saccharomyces cerevisiae expression vectors with thrombin-cleavable N- and C-terminal 6x(His) tags. Biotechnol Lett. 2003 25(4):331 - 334) and the pYes system for S. cereviseae (Invitrogen).
  • Examples of vectors for use in funghi are the pBAR series (described in Pall, M. L. and J. Brunelli. 1993. A series of six compact fungal transformation vectors containing polylinkers with unique restrictions sites. Fungal Genetics Newsletter 40: 59-61 ) .
  • the plEx plasmid based system (Merck) or the baculovirus based system (Merck) are two examples of systems useful for insect cells. Similar products are available from other companies.
  • vectors for use in insect cells include the tetracycline regulated systems pTet and pTre, the adenovirus-based system Adeno-X, the retrovirus-based system Rethro-X (all Clontech) and the pcDNA vectors (Invitrogen). Again, many more examples exist and are on the market.
  • Polypeptides according to the present invention may be expressed in mammalian cells.
  • suitable mammalian host cells include African green monkey kidney cells (Vero; ATCC CRL 1587), human embryonic kidney cells (293-HEK; ATCC CRL 1573), baby hamster kidney cells (BHK-21 , BHK-570; ATCC CRL 8544, ATCC CRL 10314), canine kidney cells (MDCK; ATCC CCL 34), Chinese hamster ovary cells (CHO-K1 ; ATCC CCL61 ; CHO DG44 [Chasin et al., Som. Cell. Molec. Genet.
  • GH1 rat pituitary cells
  • ATCC CCL82 HeLa S3 cells
  • ATCC CCL2.2 HeLa S3 cells
  • H-4-II-E rat hepatoma cells
  • COS-1 SV40-transformed monkey kidney cells
  • NIH-3T3 ATCC CRL 1658
  • the transcriptional and translational regulatory signals may be derived from viral sources, such as adenovirus, bovine papilloma virus, simian virus, or the like, in which the regulatory signals are associated with a particular gene which has a high level of expression.
  • viral sources such as adenovirus, bovine papilloma virus, simian virus, or the like, in which the regulatory signals are associated with a particular gene which has a high level of expression.
  • Suitable transcriptional and translational regulatory sequences also can be obtained from mammalian genes, such as actin, collagen, myosin, and metallothionein genes.
  • Transcriptional regulatory sequences include a promoter region sufficient to direct the initiation of RNA synthesis.
  • Suitable eukaryotic promoters include the promoter of the mouse metallothionein I gene (Hamer et al., J. Molec. Appl. Genet. 1 :273 (1982)), the TK promoter of Herpes virus (McKnight, Cell 31 :355 (1982)), the SV40 early promoter (Benoist et al., Nature 290:304 (1981 )), the Rous sarcoma virus promoter (Gorman et al., Proc. Nat'l Acad. Sci.
  • a prokaryotic promoter such as the bacteriophage T3 RNA polymerase promoter
  • a prokaryotic promoter can be used to control gene expression in mammalian cells if the prokaryotic promoter is regulated by a eukaryotic promoter (Zhou et al., MoI. Cell. Biol. 10:4529 (1990), and Kaufman et al., Nucl. Acids Res. 19:4485 (1991 )).
  • An expression vector can be introduced into host cells using a variety of standard techniques including calcium phosphate transfection, liposome-mediated transfection, microprojectile-mediated delivery, electroporation, and the like.
  • the transfected cells can be selected and propagated to provide recombinant host cells that comprise the expression vector stably integrated in the host cell genome.
  • a gene according to the present invention may thus be expressed in higher eukaryots, such as avian, fungal, insect, yeast, and plant cells.
  • one suitable selectable marker is a gene that provides resistance to the antibiotic neomycin.
  • selection is carried out in the presence of a neomycin- type drug, such as G-418 or the like.
  • Selection systems can also be used to increase the expression level of the gene of interest, a process referred to as "amplification.” Amplification is carried out by culturing transfectants in the presence of a low level of the selective agent and then increasing the amount of selective agent to select for cells that produce high levels of the products of the introduced genes.
  • a suitable amplifiable selectable marker is dihydrofolate reductase, which confers resistance to methotrexate.
  • Other drug resistance genes e.g., hygromycin resistance, multi-drug resistance, puromycin acetyltransferase
  • markers that introduce an altered phenotype such as green fluorescent polypeptide, or cell surface polypeptides such as CD4, CD8, Class I MHC, placental alkaline phosphatase may be used to sort transfected cells from untransfected cells by such means as FACS sorting or magnetic bead separation technology.
  • Polypeptides according to the present invention can also be produced by cultured mammalian cells using a viral delivery system.
  • viruses for this purpose include adenovirus, herpesvirus, vaccinia virus and adeno-associated virus (AAV).
  • Adenovirus a double-stranded DNA virus, is currently the best studied gene transfer vector for delivery of heterologous polynucleotide (for a review, see Becker et al., Meth. Cell Biol. 43:161 (1994), and Douglas and Curiel, Science & Medicine 4:44 (1997)).
  • Advantages of the adenovirus system include the accommodation of relatively large DNA inserts, the ability to grow to high-titer, the ability to infect a broad range of mammalian cell types, and flexibility that allows use with a large number of available vectors containing different promoters.
  • Adenovirus vector- infected human 293 cells ATCC Nos. CRL-1573, 45504, 45505
  • Adenovirus vector- infected human 293 cells can be grown as adherent cells or in suspension culture at relatively high cell density to produce significant amounts of polypeptide (see Gamier et al., Cytotechnol. 15:145 (1994)).
  • transgenic organisms are obtained simply by introducing the relevant
  • the baculovirus system provides an efficient means to introduce cloned genes according to the present invention into insect cells.
  • Suitable expression vectors are based upon the Autographa californica multiple nuclear polyhedrosis virus (AcMNPV), and contain well-known promoters such as Drosophila heat shock polypeptide (hsp) 70 promoter, Autographa californica nuclear polyhedrosis virus immediate-early gene promoter (ie-1 ) and the delayed early 39K promoter, baculovirus p10 promoter, and the Drosophila metallothionein promoter.
  • hsp Drosophila heat shock polypeptide
  • ie-1 Autographa californica nuclear polyhedrosis virus immediate-early gene promoter
  • baculovirus p10 promoter the Drosophila metallothionein promoter.
  • a second method of making recombinant baculovirus utilizes a transposon-based system described by Luckow (Luckow, et al., J. Virol. 67:4566 (1993)).
  • This system which utilizes transfer vectors, is sold in the BAC-to-BAC kit (Life Technologies, Rockville, Md.).
  • This system utilizes a transfer vector, PFASTBAC (Life Technologies) containing a Tn7 transposon to move the DNA encoding the polypeptide according to the present invention into a baculovirus genome maintained in E. coli as a large plasmid called a "bacmid.” See, Hill-Perkins and Possee, J. Gen. Virol.
  • transfer vectors can include an in-frame fusion with DNA encoding an epitope tag at the C- or N-terminus of the expressed polypeptide according to the present invention, for example, a GIu-GIu epitope tag (Grussenmeyer et al., Proc. Nat'l Acad. Sci. 82:7952 (1985)).
  • a transfer vector containing a gene according to the present invention is transformed into E. coli, and screened for bacmids, which contain an interrupted lacZ gene indicative of recombinant baculovirus.
  • the bacmid DNA containing the recombinant baculovirus genome is then isolated using common techniques.
  • the illustrative PFASTBAC vector can be modified to a considerable degree.
  • the polyhedrin promoter can be removed and substituted with the baculovirus basic polypeptide promoter (also known as Pcor, p6.9 or MP promoter) which is expressed earlier in the baculovirus infection, and has been shown to be advantageous for expressing secreted polypeptides (see, for example, Hill-Perkins and Possee, J. Gen. Virol. 71 :971 (1990), Bonning, et al., J. Gen. Virol. 75:1551 (1994), and
  • transfer vector constructs a short or long version of the basic polypeptide promoter can be used.
  • transfer vectors can be constructed which replace the native secretory signal sequences of polypeptides according to the present invention with secretory signal sequences derived from insect polypeptides.
  • a secretory signal sequence from Ecdysteroid Glucosyltransferase (EGT), honey bee Melittin (Invitrogen Corporation; Carlsbad, Calif.), or baculovirus gp67 (PharMingen: San Diego, Calif.) can be used in constructs to replace native secretory signal sequences.
  • the recombinant virus or bacmid is used to transfect host cells.
  • suitable insect host cells include cell lines derived from IPLB-Sf-21 , a Spodoptera frugiperda pupal ovarian cell line, such as Sf9 (ATCC CRL 171 1 ), Sf21 AE, and Sf21 (Invitrogen Corporation; San Diego, Calif.), as well as Drosophila Schneider-2 cells, and the HIGH FIVEO cell line (Invitrogen) derived from Trichoplusia ni (U.S. Pat. No. 5,300,435).
  • Sf9 ATCC CRL 171 1
  • Sf21 AE Sf21
  • Sf21 Invitrogen Corporation
  • Drosophila Schneider-2 cells Drosophila Schneider-2 cells
  • HIGH FIVEO cell line Invitrogen
  • Commercially available serum-free media can be used to grow and to maintain the cells.
  • Suitable media are Sf900 IITM (Life Technologies) or ESF 921 TM (Expression Systems) for Sf9 cells; and Ex-cellO405TM (JRH Biosciences, Lenexa, Kans.) or Express FiveOTM (Life Technologies) for T. ni cells.
  • the cells are typically grown up from an inoculation density of approximately 2 to 5 X 10 5 cells to a density of 1 to 2 X 10 6 cells at which time a recombinant viral stock is added at a multiplicity of infection (MOI) of 0.1 to 10, more typically near 3.
  • MOI multiplicity of infection
  • yeast cells can also be used to express the genes described herein.
  • Yeast species of particular interest in this regard include Saccharomyces cerevisiae, Pichia pastoris, and Pichia methanolica.
  • Suitable promoters for expression in yeast include promoters from GAL1 (galactose), PGK (phosphoglycerate kinase), ADH (alcohol dehydrogenase), AOX1 (alcohol oxidase), HIS4 (histidinol dehydrogenase), and the like.
  • GAL1 galactose
  • PGK phosphoglycerate kinase
  • ADH alcohol dehydrogenase
  • AOX1 alcohol oxidase
  • HIS4 histidinol dehydrogenase
  • vectors include Ylp-based vectors, such as Ylp5, YRp vectors, such as YRp17, YEp vectors such as YEp13 and YCp vectors, such as YCp19.
  • Methods for transforming S. cerevisiae cells with exogenous DNA and producing recombinant polypeptides therefrom are disclosed by, for example, Kawasaki, U.S. Pat. No. 4,599,31 1 , Kawasaki et al., U.S. Pat. No. 4,931 ,373, Brake, U.S. Pat. No. 4,870,008, Welch et al., U.S. Pat. No. 5,037,743, and Murray et al., U.S. Pat. No.
  • Transformed cells are selected by phenotype determined by the selectable marker, commonly drug resistance or the ability to grow in the absence of a particular nutrient (e.g., leucine).
  • a suitable vector system for use in Saccharomyces cerevisiae is the POT1 vector system disclosed by Kawasaki et al. (U.S. Pat. No. 4,931 ,373), which allows transformed cells to be selected by growth in glucose-containing media. Additional suitable promoters and terminators for use in yeast include those from glycolytic enzyme genes (see, e.g., Kawasaki, U.S. Pat. No. 4,599,31 1 , Kingsman et al., U.S. Pat. No.
  • Transformation systems for other yeasts including Hansenula polymorpha, Schizosaccharomyces pombe, Kluyveromyces lactis, Kluyveromyces fragilis, Ustilago maydis, Pichia pastoris, Pichia methanolica, Pichia guillermondii and Candida maltosa are known in the art. See, for example, Gleeson et al., J. Gen. Microbiol. 132:3459
  • Pichia methanolica as host for the production of recombinant polypeptides is disclosed by Raymond, U.S. Pat. No. 5,716,808, Raymond, U.S. Pat. No. 5,736,383, Raymond et al., Yeast 14:1 1 23 (1998), and in international publication Nos. WO 97/17450, WO 97/17451 , WO 98/02536, and WO 98/02565.
  • DNA molecules for use in transforming P. methanolica will commonly be prepared as double-stranded, circular plasmids, which can be linearized prior to transformation. For polypeptide production in P.
  • the promoter and terminator in the plasmid can be that of a P. methanolica gene, such as a P. methanolica alcohol utilization gene (AUG1 or AUG2).
  • P. methanolica alcohol utilization gene AUG2
  • Other useful promoters include those of the dihydroxyacetone synthase (DHAS), formate dehydrogenase (FMD), and catalase (CAT) genes.
  • DHAS dihydroxyacetone synthase
  • FMD formate dehydrogenase
  • CAT catalase
  • a suitable selectable marker for use in Pichia methanolica is a P. methanolica ADE2 gene, which encodes phosphoribosyl-5-aminoimidazole carboxylase (AIRC; EC
  • methanolica cells can be transformed by electroporation using an exponentially decaying, pulsed electric field having a field strength of from 2.5 to 4.5 kV/cm, preferably about 3.75 kV/cm, and a time constant (t) of from 1 to 40 milliseconds, most preferably about 20 milliseconds.
  • Expression vectors can also be introduced into plant protoplasts, intact plant tissues, or isolated plant cells.
  • Methods for introducing expression vectors into plant tissue include the direct infection or co-cultivation of plant tissue with Agrobacterium tumefaciens, microprojectile-mediated delivery, DNA injection, electroporation, and the like. See, for example, Horsch et al., Science 227:1229 (1985), Klein et al., Biotechnology 10:268 (1992), and Miki et al., "Procedures for Introducing Foreign DNA into Plants," in Methods in Plant Molecular Biology and Biotechnology, Glick et al. (eds.), pages 67 88 (CRC Press, 1993).
  • genes according to the present invention can be expressed in prokaryotic host cells.
  • Suitable promoters that can be used to express polypeptides according to the present invention in a prokaryotic host are well-known to those of skill in the art and include promoters capable of recognizing the T4, T3, Sp6 and T7 polymerases, the P R and P L promoters of bacteriophage lambda, the trp, recA, heat shock, lacUV5, tac, Ipp- lacSpr, phoA, and lacZ promoters of E. coli, promoters of B.
  • subtilis the promoters of the bacteriophages of Bacillus, Streptomyces promoters, the int promoter of bacteriophage lambda, the bla promoter of pBR322, and the CAT promoter of the chloramphenicol acetyl transferase gene.
  • Prokaryotic promoters have been reviewed by Glick, J. Ind. Microbiol. 1 :277 (1987), Watson et al., Molecular Biology of the Gene, 4th Ed. (Benjamin Cummins 1987), and by Ausubel et al. (1995).
  • Suitable prokaryotic hosts include E. coli and Bacillus subtilus.
  • Suitable strains of E. coli include BL21 (DE3), BL21 (DE3)pLysS, BL21 (DE3)pLysE, DH1 , DH4I, DH5, DH5I, DH5IF, DH5IMCR, DH10B, DH10B/p3, DH1 1 S, C600, HB101 , JM101 , JM105, JM109,
  • Suitable strains of Bacillus subtilus include BR151 , YB886, MM 19, MM 20, and B170 (see, for example, Hardy, "Bacillus Cloning Methods," in DNA Cloning: A Practical Approach, Glover (ed.) (IRL Press 1985)).
  • a polypeptide according to the present invention in bacteria such as E.
  • the polypeptide may be retained in the cytoplasm, typically as insoluble granules, or may be directed to the periplasmic space by a bacterial secretion sequence.
  • the cells are lysed, and the granules are recovered and denatured using, for example, guanidine isothiocyanate or urea.
  • the denatured polypeptide can then be refolded and dimerized by diluting the denaturant, such as by dialysis against a solution of urea and a combination of reduced and oxidized glutathione, followed by dialysis against a buffered saline solution.
  • the polypeptide can be recovered from the periplasmic space in a soluble and functional form by disrupting the cells (by, for example, sonication or osmotic shock) to release the contents of the periplasmic space and recovering the polypeptide, thereby obviating the need for denaturation and refolding.
  • polypeptides of the present invention can be synthesized by exclusive solid phase synthesis, partial solid phase methods, fragment condensation or classical solution synthesis. These synthesis methods are well-known to those of skill in the art (see, for example, Merrifield, J. Am. Chem. Soc. 85:2149 (1963), Stewart et al., “Solid Phase Peptide Synthesis” (2nd Edition), (Pierce Chemical Co. 1984), Bayer and Rapp, Chem. Pept. Prot.
  • compositions comprising a peptide or polypeptide described herein.
  • Such compositions can further comprise a carrier.
  • the carrier can be a conventional organic or inorganic carrier. Examples of carriers include water, buffer solution, alcohol, propylene glycol, macrogol, sesame oil, corn oil, and the like.
  • polypeptides of the present invention can be purified to at least about 80% purity, to at least about 90% purity, to at least about 95% purity, or even greater than 95% purity with respect to contaminating macromolecules, particularly other polypeptides and polynucleotides, and free of infectious and pyrogenic agents.
  • the polypeptides of the present invention can also be purified to a pharmaceutically pure state, which is greater than 99.9% pure.
  • a purified polypeptide is substantially free of other polypeptides, particularly other polypeptides of animal origin.
  • Fractionation and/or conventional purification methods can be used to obtain preparations of polypeptides according to the present invention purified from natural sources, and recombinant polypeptides according to the present invention and fusion polypeptides according to the present invention purified from recombinant host cells.
  • ammonium sulfate precipitation and acid or chaotrope extraction may be used for fractionation of samples.
  • Exemplary purification steps may include hydroxyapatite, size exclusion, FPLC and reverse-phase high performance liquid chromatography. Suitable chromatographic media include derivatized dextrans, agarose, cellulose, polyacrylamide, specialty silicas, and the like. PEI, DEAE, QAE and Q derivatives are preferred.
  • Exemplary chromatographic media include those media derivatized with phenyl, butyl, or octyl groups, such as Phenyl-Sepharose FF (Pharmacia), Toyopearl butyl 650 (Toso Haas, Montgomeryville, Pa.), Octyl-Sepharose (Pharmacia) and the like; or polyacrylic resins, such as Amberchrom CG 71 (Toso Haas) and the like.
  • Suitable solid supports include glass beads, silica-based resins, cellulosic resins, agarose beads, cross-linked agarose beads, polystyrene beads, cross-linked polyacrylamide resins and the like that are insoluble under the conditions in which they are to be used. These supports may be modified with reactive groups that allow attachment of polypeptides by amino groups, carboxyl groups, sulfhydryl groups, hydroxyl groups and/or carbohydrate moieties.
  • Examples of coupling chemistries include cyanogen bromide activation, N- hydroxysuccinimide activation, epoxide activation, sulfhydryl activation, hydrazide activation, and carboxyl and amino derivatives for carbodiimide coupling chemistries.
  • polypeptides according to the present invention can be devised by those of skill in the art.
  • specific antibodies recognising polypeptides according to the present invention and fragments thereof, obtained as described below can be used to isolate large quantities of polypeptide by immunoaffinity purification.
  • the polypeptides of the present invention can also be isolated by exploitation of particular properties.
  • immobilized metal ion adsorption (IMAC) chromatography can be used to purify histidine-rich polypeptides, including those comprising polyhistidine tags. Briefly, a gel is first charged with divalent metal ions to form a chelate (Sulkowski, Trends in Biochem. 3:1 (1985)). Histidine-rich polypeptides will be adsorbed to this matrix with differing affinities, depending upon the metal ion used, and will be eluted by competitive elution, lowering the pH, or use of strong chelating agents.
  • IMAC immobilized metal ion adsorption
  • a fusion of the polypeptide of interest and an affinity tag may be constructed to facilitate purification.
  • an affinity tag e.g., maltose- binding polypeptide, an immunoglobulin domain
  • Polypeptides and fragments thereof according to the present invention may also be prepared through chemical synthesis, as described above.
  • Polypeptides according to the present invention may be monomers or multimers; glycosylated or non- glycosylated; pegylated or non-pegylated; and may or may not include an initial methionine amino acid residue.
  • Antibodies to an ice-binding polypeptide according to the present invention, or a fragment thereof can be obtained, for example, by using as an antigen the product produced from an expression vector comprising a gene according to the present invention in a suitable host organism, or by using a polypeptide according to the present invention isolated from a natural source or synthesised using any conventional solid phase synthesis strategy.
  • Particularly useful antibodies "bind specifically" with a polypeptide according to the present invention.
  • Antibodies are considered to be specifically binding if the antibodies exhibit at least one of the following two properties: (1 ) antibodies bind to a polypeptide according to the present invention with a threshold level of binding activity, and (2) antibodies do not significantly cross-react with polypeptides which are related to a polypeptide according to the present invention as defined herein below.
  • antibodies specifically bind if they bind to a polypeptide, peptide or epitope with a binding affinity (K a ) of 10 6 M -1 or greater, preferably 10 7 M -1 or greater, more preferably 10 8 M -1 or greater, and most preferably 10 9 M "1 or greater.
  • K a binding affinity
  • the binding affinity of an antibody can be readily determined by one of ordinary skill in the art, for example, by Scatchard analysis (Scatchard, Ann. NY Acad. Sci. 51 :660 (1949)).
  • antibodies do not significantly cross-react with related polypeptide molecules, for example, if they detect polypeptides according to the present invention, but do not detect known polypeptides applied in similar or identical amounts in a standard Western blot analysis.
  • Antibodies can be produced using antigenic epitope-bearing peptides or polypeptides according to the present invention.
  • Antigenic epitope-bearing peptides and polypeptides of the present invention preferably contain a sequence of at least four, or between 15 to about 30 amino acids contained within any of SEQ ID NOs:5 to 12.
  • peptides or polypeptides comprising a larger portion of an amino acid sequence of the invention, containing from 30 to 50 amino acids, or any length up to and including the entire amino acid sequence of a polypeptide according to the invention, also are useful for inducing antibodies that bind with polypeptides according to the present invention.
  • amino acid sequence of the epitope- bearing peptide is selected to provide substantial solubility in aqueous solvents (i.e., the sequence includes relatively hydrophilic residues, while hydrophobic residues are preferably avoided). Moreover, amino acid sequences containing proline residues may be also be desirable for antibody production.
  • potential antigenic sites in polypeptides according to the present invention can be identified using the Jameson-Wolf method, Jameson and Wolf, CABIOS 4:181 , (1988), as implemented by the PROTEAN program (version 3.14) of LASERGENE (DNASTAR; Madison, Wis.). Default parameters were used in this analysis.
  • Polyclonal antibodies to recombinant polypeptide or to isolated from natural sources can be prepared using methods well-known to those of skill in the art. See, for example, Green et al., "Production of Polyclonal Antisera,” in Immunochemical Protocols (Manson, ed.), pages 1 to 5 (Humana Press 1992), and Williams et al., "Expression of foreign polypeptides in E. coli using plasmid vectors and purification of specific polyclonal antibodies," in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 15 (Oxford University Press 1995).
  • the immunogenicity of a polypeptide can be increased through the use of an adjuvant, such as alum (aluminum hydroxide) or Freund's complete or incomplete adjuvant.
  • Polypeptides useful for immunization also include fusion polypeptides, such as fusions of or a portion thereof with an immunoglobulin polypeptide or with maltose binding polypeptide.
  • the polypeptide immunogen may be a full-length molecule or a portion thereof. If the polypeptide portion is "hapten-like,” such portion may be advantageously joined or linked to a macromolecular carrier (such as keyhole limpet hemocyanin (KLH), bovine serum albumin (BSA) or tetanus toxoid) for immunization.
  • KLH keyhole limpet hemocyanin
  • BSA bovine serum albumin
  • tetanus toxoid tetanus toxoid
  • polyclonal antibodies are typically raised in animals such as horses, cows, dogs, chicken, rats, mice, rabbits, guinea pigs, goats, or sheep
  • an antibody specific for a polypeptides according to the present invention may also be derived from a subhuman primate antibody.
  • General techniques for raising diagnostically and therapeutically useful antibodies in baboons may be found, for example, in Goldenberg et al., international patent publication No. WO 91/1 1465, and in Losman et al., Int. J. Cancer 46:310 (1990).
  • monoclonal antibodies specific for a polypeptides according to the present invention can be generated.
  • Rodent monoclonal antibodies to specific antigens may be obtained by methods known to those skilled in the art (see, for example, Kohler et al., Nature 256:495 (1975), Coligan et al. (eds.), Current Protocols in Immunology, Vol. 1 , pages 2.5.1 2.6.7 (John Wiley & Sons 1991 ) ["Coligan”], Picksley et al., "Production of monoclonal antibodies against polypeptides expressed in E. coli," in DNA Cloning 2: Expression Systems, 2nd Edition, Glover et al. (eds.), page 93 (Oxford University Press 1995)).
  • monoclonal antibodies can be obtained by injecting mice with a composition comprising a gene product, verifying the presence of antibody production by removing a serum sample, removing the spleen to obtain B- lymphocytes, fusing the B- lymphocytes with myeloma cells to produce hybridomas, cloning the hybridomas, selecting positive clones which produce antibodies to the antigen, culturing the clones that produce antibodies to the antigen, and isolating the antibodies from the hybridoma cultures.
  • an antibody specific for polypeptides according to the present invention of the present invention may be derived from a human monoclonal antibody.
  • Human monoclonal antibodies are obtained from transgenic mice that have been engineered to produce specific human antibodies in response to antigenic challenge.
  • elements of the human heavy and light chain locus are introduced into strains of mice derived from embryonic stem cell lines that contain targeted disruptions of the endogenous heavy chain and light chain loci.
  • the transgenic mice can synthesize human antibodies specific for human antigens, and the mice can be used to produce human antibody-secreting hybridomas.
  • Methods for obtaining human antibodies from transgenic mice are described, for example, by Green et al., Nature Genet. 7:13 (1994), Lonberg et al., Nature 368:856 (1994), and Taylor et al., Int. Immun. 6:579 (1994).
  • Monoclonal antibodies can be isolated and purified from hybridoma cultures by a variety of well-established techniques. Such isolation techniques include affinity chromatography with polypeptide-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography (see, for example, Coligan at pages 2.7.1 2.7.12 and pages 2.9.1 2.9.3; Baines et al., "Purification of Immunoglobulin G (IgG),” in Methods in Molecular Biology, Vol. 10, pages 79 104 (The Humana Press, Inc. 1992)).
  • isolation techniques include affinity chromatography with polypeptide-A Sepharose, size-exclusion chromatography, and ion-exchange chromatography (see, for example, Coligan at pages 2.7.1 2.7.12 and pages 2.9.1 2.9.3; Baines et al., "Purification of Immunoglobulin G (IgG),” in Methods in Molecular Biology, Vol. 10, pages 79 104 (The Humana Press, Inc
  • antibody fragments can be obtained, for example, by proteolytic hydrolysis of the antibody.
  • Antibody fragments can be obtained by pepsin or papain digestion of whole antibodies by conventional methods.
  • antibody fragments can be produced by enzymatic cleavage of antibodies with pepsin to provide a 5S fragment denoted F(ab') 2 .
  • This fragment can be further cleaved using a thiol reducing agent to produce 3.5S Fab' monovalent fragments.
  • the cleavage reaction can be performed using a blocking group for the sulfhydryl groups that result from cleavage of disulfide linkages.
  • an enzymatic cleavage using pepsin produces two monovalent Fab fragments and an F c fragment directly.
  • These methods are described, for example, by Goldenberg, U.S. Pat. No. 4,331 ,647, Nisonoff et al., Arch Biochem. Biophys. 89:230 (1960), Porter, Biochem. J. 73:1 19 (1959), Edelman et al. and Coligan, both in Methods in Enzymology Vol. 1 , (Academic Press 1967).
  • cleaving antibodies such as separation of heavy chains to form monovalent light-heavy chain fragments, further cleavage of fragments, or other enzymatic, chemical or genetic techniques may also be used, so long as the fragments bind to the antigen that is recognized by the intact antibody.
  • Fv fragments comprise an association of V H and V L chains. This association can be noncovalent, as described by lnbar et al., Proc. Nat'l Acad. Sci. USA 69:2659 (1972).
  • the variable chains can be linked by an intermolecular disulfide bond or cross-linked by chemicals such as glutaraldehyde (see, for example, Sandhu, Crit. Rev. Biotech. 12:437 (1992)).
  • the Fv fragments may comprise V H and V L chains, which are connected by a peptide linker.
  • scFv single-chain antigen binding polypeptides
  • a scFV can be obtained by exposing lymphocytes to polypeptide in vitro, and selecting antibody display libraries in phage or similar vectors (for instance, through use of immobilized or labeled polypeptide or peptide).
  • Genes encoding polypeptides having potential polypeptide binding domains can be obtained by screening random peptide libraries displayed on phage (phage display) or on bacteria, such as E. coli.
  • Nucleotide sequences encoding the polypeptides can be obtained in a number of ways, such as through random mutagenesis and random polynucleotide synthesis.
  • random peptide display libraries can be used to screen for peptides, which interact with a known target which can be a polypeptide or polypeptide, such as a ligand or receptor, a biological or synthetic macromolecule, or organic or inorganic substances.
  • a known target which can be a polypeptide or polypeptide, such as a ligand or receptor, a biological or synthetic macromolecule, or organic or inorganic substances.
  • Techniques for creating and screening such random peptide display libraries are known in the art (Ladner et al., U.S. Pat. No. 5,223,409, Ladner et al., U.S. Pat. No. 4,946,778, Ladner et al., U.S. Pat. No. 5,403,484, Ladner et al., U.S. Pat. No.
  • Random peptide display libraries can be screened using the sequences disclosed herein to identify polypeptides which bind to .
  • CDR peptides (“minimal recognition units") can be obtained by constructing genes encoding the CDR of an antibody of interest. Such genes are prepared, for example, by using the polymerase chain reaction to synthesize the variable region from RNA of antibody-producing cells (see, for example, Larrick et al., Methods: A Companion to Methods in Enzymology 2:106 (1991 ), Courtenay-Luck, "Genetic Manipulation of Monoclonal Antibodies," in Monoclonal Antibodies: Production, Engineering and Clinical Application, Ritter et al.
  • an antibody specific for a polypeptide according to the present invention may be derived from a "humanized" monoclonal antibody.
  • Humanized monoclonal antibodies are produced by transferring mouse complementary determining regions from heavy and light variable chains of the mouse immunoglobulin into a human variable domain. Typical residues of human antibodies are then substituted in the framework regions of the murine counterparts.
  • the use of antibody components derived from humanized monoclonal antibodies obviates potential problems associated with the immunogenicity of murine constant regions. General techniques for cloning murine immunoglobulin variable domains are described, for example, by Orlandi et al., Proc. Nat'l Acad. Sci. USA 86:3833 (1989).
  • Polyclonal anti-idiotype antibodies can be prepared by immunizing animals with antibodies or antibody fragments specific for a polypeptide according to the present invention, using standard techniques. See, for example, Green et al., "Production of Polyclonal Antisera,” in Methods In Molecular Biology: Immunochemical Protocols, Manson (ed.), pages 1 12 (Humana Press 1992). Also, see Coligan at pages 241 to 247.
  • monoclonal anti-idiotype antibodies can be prepared using antibodies or antibody fragments specific for a polypeptide according to the present invention as immunogens with the techniques, described above.
  • humanized anti-idiotype antibodies or subhuman primate anti-idiotype antibodies can be prepared using the above-described techniques.
  • the host cells which may comprise a polypeptide according to the present invention can be examplified e.g. by animal cells, mammalian host cells, insect cells, fish cells, fungal cells, yeast cells, bacterial cells and plant cells.
  • polynucleotide constructs capable of introduction to and/or expression in the ultimate target expressing cell will be suitable for replication in a unicellular or multicellular host, such as yeast or bacteria, but may also be intended for introduction and integration within the genome of cultured mammalian or other eukaryotic cell lines, in particular, plants, polynucleotide constructs prepared for introduction into bacteria or yeast will include a replication system recognized by the host, the polynucleotide fragment encoding the desired anti-freeze polypeptide product, transcriptional and translational initiation regulatory sequences joined to the 5'-end of the anti-freeze polypeptide encoding polynucleotide sequence, and transcriptional and translational termination regulatory sequences joined to the 3'-end of the sequence.
  • the transcriptional regulatory sequences will include a heterologous promoter which is recognized by the host.
  • available expression vectors which include the replication system and transcriptional and translational regulatory sequences together with an insertion site for the anti-freeze polypeptide encoding sequence may be employed.
  • the gene will include any polynucleotide segment which contains a coding sequence for anti-freeze polypeptide. Normally, the gene will include the coding sequence plus the upstream and downstream associated sequences, particularly any enhancer, promoter, ribosome binding site or transcription initiation markers. Downstream segments may also be important for message polyadenylation and processing, and thus are also contemplated in the usual instance.
  • the introduction of genes into cells or groups of cells for expression is another method for generally introducing the polypeptides into the sample of interest.
  • the end product of the transformation is also included as the product of this invention, and the term "transformed cell" will include the actual cell transformed, and all progeny of that cell. In the typical case, the final organism will contain cells, each of which will contain the gene.
  • yeast host cells suitable for use in accordance with the present invention include yeasts from the Family of Saccharomycetaceae, including members of the genera Saccharomyces and Candida. Preferred examples include, but are not limited to, Saccharomyces fragilis, Saccharomyces cervisae, Saccharomyces lactis, Candida pseudotropicalis.
  • Bacteria Bacterial cells are useful as host cells according to the present invention for the production of the polypeptides according to the present invention.
  • Bacteria e.g. Lactobacillus as well as many yeasts and molds, have been used for thousands of years in the preparation of fermented foods, such as e.g. cheese, pickles, soy sauce, sauerkraut, vinegar, wine and yoghurt.
  • Anti-freeze polypeptides according to the present invention are useful in maintaining the viability of the microorganisms used to prepare such foods, as well as in the preparation of prebiotic and probiotic edible compositions, including animal feed compositions and foods for human consumption.
  • Examples of preferred bacteria relevant to the present invention and suitable as host cells in accordance with the present invention are, for example, Escherichia coli, Streptococcus cremoris, Streptococcus lactis, Streptococcus thermophilus, Leuconostoc citrovorum, Leuconostoc mesenteroides, Lactobacillus acidophilus, Lactobacillus lactis, Lactobacillus bulgaricus, Bifidobacterium bifidum, Bifidobacterium breve, Lactobacillus delbrueckii ssp.
  • Bacteria can also be used as substitutes for pesticides in a biological pest control programme.
  • the invention in one embodiment is particularly well suited for such applications and provide recombinant microrganisms harbouring polynucleotides according to the invention and producing polypeptides according to the invention capable of being used as environmentally friendly, biological pesticides.
  • Bacillus thurmgiensis a Gram-positive, soil dwelling bacterium.
  • Further non-limiting examples of bacterial host cells suitable for use in accordance with the present invention include Gram-positive bacteria and Gram-negative bacteria.
  • Preferred bacterial cells can also be selected from the groups consisting of Gram- positive cocci, Gram-positive bacilli, Gram-negative cocci and Gram-negative bacilli.
  • Examples of bacterial host cells suitable for use in accordance with the present invention include, but is not limited to, bacteria selected from the following genera: Acaricomes, Acetitomaculum, Acetivibrio, Acetobacter, Acetobacterium,
  • Actinocatenispora Actinocorallia, Actinokineospora, Actinomadura, Actinomyces, Actinoplanes, Actinopolyspora, Actinopycnidium, Actinospica, Actinosporangium, Actinostreptospora, Actinosynnema, Actinotalea, Actinotelluria, Adhaeribacter, Aequorivita, Aerobacter, Aerococcus, Aerococcus-like Organism, Aeromicrobium, Aeromonas, Aestuariibacter, Afipia, Agarbacterium, Aggregatibacter, Agitococcus,
  • Asticcacaulis Astrosporangium, Atopobium, Atopococcus, Atopostipes, Aurantimonas, Aureobacterium, Avibacterium, Axonoporis, Azoarcus, Azohydromonas, Azomonas, Azorhizobium, Azorhizophilus, Azospira, Azospirillum, Azotobacter Bacillus, Bacteriovorax, Bacterium, Bacteroides, Balnearium, Balneatrix, Barnesiella, Bartonella, Bdellovibrio, Beggiatoa, Beijerinckia, BeIMeIIa, Belnapia, Beneckea, Bergeriella,
  • Betabacterium Beutenbergia, Bifidobacterium, Bilophila, Blastobacter, Blastochloris, Blastococcus, Blastomonas, Blastopirellula, Bogoriella, Bordetella, Borrelia, Bosea, Brachybacterium, Brachymonas, Brachyspira, Brackiella, Bradyrhizobium, Branhamella, Brenneria, Brevibacillus, Brevibacterium, Brevigemma, Brevundimonas, Brochothrix, Brucella, Bryantella, Budvicia, Bulleidia, Burkholderia, Buttiauxella, Butyribacterium, Butyrivibrio, Byssovorax, Caenibacterium, Caldanaerobacter, Caldicellulosiruptor, Caldilinea, Caldithrix, Caldocellum, Caloramator, Caloranaerobacter, Caminibacillus, Caminibacter,
  • Desulfonatronum Desulfonauticus, Desulfonema, Desulfonispora, Desulfopila, Desulforegula, Desulforhabdus, Desulforhopalus, Desulfosarcina, Desulfospira, Desulfosporosinus, Desulfotalea, Desulfothermus, Desulfotignum, Desulfotomaculum, Desulfovermiculus, Desulfovibrio, Desulfovirga, Desulfovirgula, Desulfurella, Desulfurobacterium, Desulfuromonas, Desulfuromusa, Dethiosulfovibrio, Devosia, Dialister, Diaphorobacter, Dichelobacter, Dichotomicrobium, Dickeya, Dictyoglomus, Dietzia, Diplococcus, Dokdoa, Dokdonella, Dokdonia, Dolosicoccus, Donghaeana
  • Geopsychrobacter Georgenia, Geosinus, Geospirillum, Geothermobacter, Geothrix, Geovibrio, Giesbergeria, Gillisia, Glaciecola, Globicatella, Gluconacetobacter, Gluconoacetobacter, Gluconobacter, Glycomyces, Goodfellowia, Gordona, Gordonia, Gracilibacillus, Gracilibacter, Granulicatella, Granulobacter, Grimontia, Group Ii D, Guggenheimella, Gulosibacter, Haemophilus, Hafnia, Hahella, Halanaerobacter,
  • Halanaerobium Haliangium, Haliscomenobacter, Haloactinomyces, Haloanaerobacter, Haloanaerobium, Halobacillus, Halobacteroides, Halocella, Halochromatium, Halococcus, Halolactibacillus, Halomonas, Halonatronum, Halorhodospira, Halothermothrix, Halothiobacillus, Helcococcus, Helicobacter, Heliobacillus, Heliobacterium, Heliophilum, Heliorestis, Herbaspirillum, Herbidospora, Herminiimonas, Herpetosiphon, Hespellia, Hippea, Hirschia, Hoeflea, Holdemania, Holophaga, Hongiella, Hordeomyces, Hyalangium, Hydrocarboniphaga, Hydrogenivirga, Hydrogenobacter, Hydrogenobaculum, Hydrogenomonas, Hydrogenophaga, Hydrogenophil
  • Leisingera Leminorella, Lentibacillus, Lentzea, Leptospirillum, Leptothrix, Leptotrichia, Leucobacter, Leuconostoc, Leucothrix, Levilinea, Levinea, Limnobacter, List, Listeria, Listonella, Loktanella, Lonepinella, Longispora, Lophomonas, Luteibacter, Luteimonas, Luteococcus, Lysobacter, Macrococcus, Macromonas, Magnetospirillum, Mahella, Malikia, Malonomonas, Manjusharmella, Mannheimia, Maribacter, Maricaulis, Marichromatium, Marinibacillus, Marinilabilia, Marinilactibacillus, Marinithermus, Marinitoga, Marinobacter, Marinobacterium, Marinococcus, Marinomonas, Marinospirillum, Marinovum, Marmoricola, Massilia, Mechercharimy
  • Microstreptospora Microtetraspora, Microvirgula, Millisia, Mima, Mitsuokella, Mobiluncus, Modestobacter, Moellerella, Mogibacterium, Moorella, Moraxella, Moraxella (Branhamella), Moraxella (Moraxella), Morganella, Moritella, Muricauda, Muricoccus, Myceligenerans, Mycetocola, Mycobacterium, Mycoplana, Myroides, Myxobacter, Myxococcus, , Nakamurella, Nannocystis, Natroniella, Natronincola, Nautilia, Naxibacter, Neisseria, Nereida, Nesterenkonia, Nevskia, Nicoletella, Nih Group 12, Nitratifractor, Nitratireductor, Nitratiruptor, Nitrobacter, Nocardia, Nocardioides, Nocardiopsis, Nonomuraea, Novosphingobium, Obesumbacterium, Ocean
  • Octadecabacter Odontomyces, Oenococcus, Oerskovia, Oleiphilus, Oleispira, Oligella, Oligotropha, Olsenella, Opitutus, Orenia, Oribacterium, Ornithinicoccus, Ornithinimicrobium, Ornithobacterium, Oryzihumus, Ottowia, Oxalicibacterium, Oxalobacter, Oxalophagus, Oxobacter, Paenibacillus, Paludibacter, Pandoraea, Pannonibacter, Pantoea, Papillibacter, Parabacteroides, Paracoccus, Paracolobactrum, Paralactobacillus, Paraliobacillus, Parascardovia, Parasporobacterium, Parvibaculum, Parvimonas, Parvopolyspora, Pasteurella, Pasteuria, Patulibacter, Paucibacter, Paucimonas, Paucisalibacillus, Pectinatus, Pectobacter
  • Planomonospora Planosporangium, Planotetraspora, Plantibacter, Pleomorphomonas, Plesiocystis, Plesiomonas, Podangium, Polaribacter, Polaromonas, Polyangium, Polymorphospora, Pontibacillus, Porphyrobacter, Porphyromonas, Pragia, Prauserella, Prevotella, Proactinomyces, Promicromonospora, Promyxobacterium, Propionibacterium, Propionicimonas, Propioniferax, Propionigenium,
  • Rubrobacter Ruegeria, Ruminobacter, Ruminococcus, Runella, , Saccharibacter, Saccharococcus, Saccharomonospora, Saccharophagus, Saccharopolyspora, Saccharothrix, Sagittula, Salana, Salegentibacter, Salinibacter, Salinibacterium, Salinicoccus, Salinimonas, Salinispora, Salinivibrio, Salinospora, Salipiger, Salmonella, Samsonia, Sanguibacter, Saprospira, Sarcina, Sarraceniospora, Scardovia,
  • Xenorhabdus Xylanibacter, Xylanibacterium, Xylanimicrobium, Xylanimonas, XyIeIIa, Xylophilus, Yania, Yersinia, Yokenella, , Zavarzinia, Zimmermannella, Zobellia, Zoogloea, Zooshikella, Zymobacter, Zymobacterium, Zymomonas and Zymophilus.
  • the use of the polynucleotides and polypeptides according to the present invention in plant host cells and other transgenic organisms can prevent the loss of valuable crops when the climatic conditions are not optimal for the production of the crops.
  • the present invention provides novel and innovative, transgenic plants and crops capable of sustaining climatic conditions which cannot be withstood by state-of- the-art plants and crops.
  • crops in the form of plant host cells according to the present invention comprising polynucleotides and producing polypeptides according to the present invention are are: grapes, oilseed plants, such as canola, grains (oats, barley, rye etc.), citrus fruits, and sugar cane.
  • the invention is also directed to trans-genic fruits and vegetables comprising the polypeptides according to the present invention.
  • the trans-genic fruits and vegetables comprising the polypeptides according to the present invention are capable of withstanding cooler temperatures e.g. during storage and/or transport. Examples include strawberries, blueberries, raspberries, citrus fruits, bananas, grapes, kiwis, peaches, pineapples, plums, cherries, tomatoes and mangoes.
  • Flowers comprising polypeptides according to the present invention Frozen flowers crossmplated in accordance with the present invention comprised e.g. tulips, roses, lilies.
  • Examples of fish suitable for the invention are Albacore Tuna (Thunnus alalunga),
  • Anti-freeze polypeptides according to the present invention can be used to treat frozen foods or foods to be frozen in order to prevent re-crystallization.
  • foods for treatment with the invention include, but is not limited to: Ice cream, frozen yoghurt, soups, puddings, sorbets, ice cream bars, frozen desserts e.g. custards, puddings etc and other liquids or semi-liquids for freezing.
  • Frozen vegetables such as celery, potatoes, asparagus, peas, carots, beans, broccoli, sweet corn, spinach, haricots verts etc. is also encompassed by the present invention.
  • the polypeptides according to the present invention may also affect the formation of lactose crystals. Hence, without being bound by any specific theory, it is believed that the polypeptides according to the present invention inhibit the crystallisation and/or growth of lactose crystals. It is well known that during freezing of ice creams the content of all ingredients (including lactose) is increasingly concentrated except for the content of liquid water, which is decreasing. When the content of lactose reaches a certain level, lactose crystals start to crystallize. This crystallisation is a slow process, which takes place at -20°C. Typically, ice creams are stored at about -20°C.
  • Frozen or refrigerated foods are now a mainstay of the human diet in developed countries. Thus extensive research has and is being carried out by food scientists to ensure high quality products for the consumers. This is particularly true with regard to frozen vegetables and frozen deserts such as ice cream and yogurt.
  • Frozen deserts such as ice cream or yogurt are generally eaten in the frozen state.
  • Texture is to a large extent governed by the size of the ice crystals.
  • Producers of these frozen deserts have gone to considerable effort and expense to ensure smooth textured products.
  • the growth of ice crystals during frozen storage is known as recrystallization. This problem is particularly common when the frozen storage conditions are less than ideal, such as during transportation or storage in modern frost-free home freezers. After a relatively short period of time at above-zero temperatures (i.e., above 0°C), or even at sustained freezing temperatures, frozen foods can become less desirable or even unsuitable for human consumption due to the ice recrystallization process.
  • An ideal method of incorporating anti-freeze polypeptides into frozen fermented food products is to have the organism responsible for the fermentation process produce the anti-freeze polypeptides while fermenting the food.
  • the present invention embraces methods for preparing a frozen fermented food product.
  • This method comprises the steps of (a) contacting a food product with a microorganism that is capable of secreting a polypeptide according to the present invention, wherein the microorganism is capable of fermenting the food product to produce the fermented food product, (b) incubating the food product with the microorganism under conditions in which fermentation takes place so that a fermented food product is produced having the anti-freeze polypeptide present in an amount effective at inhibiting recrystallization of the product; and (c) freezing the fermented food product at a temperature below -5°C, so as to produce a frozen fermented food product.
  • the food product may be a dairy product (e.g., milk) which can be fermented to produce yogurt, buttermilk or cheese.
  • dairy product e.g., milk
  • the microorganism of the invention is usually a bacterium (e.g., Lactobacillus bulgaricus; Streptococcus cremoris, Streptococcus lactis; Bifidobacterium bifidum, Bifidobacterium longum) but may also be a fungus such as a yeast (e.g., Saccharomyces fragilis, Saccharomyces cerevisiae, Saccharomyces lactis, and others). According to the invention these microorganisms are genetically engineered so that they are capable of secreting a polypeptide according to the present invention.
  • a bacterium e.g., Lactobacillus bulgaricus; Streptococcus cremoris, Streptococcus lactis; Bifidobacterium bifidum, Bifidobacterium longum
  • yeast e.g., Saccharomyces fragilis, Saccharomyces cerevisiae,
  • the invention comprises incubating milk with bacterial species Lactobacillus balgaricus and Streptococcus lactis that are capable of fermenting milk to produce yogurt and capable of secreting anti-freeze polypeptides; incubating the bacteria and milk under conditions that produce yogurt; and freezing the yogurt at a temperature below -5°C, so as to produce frozen yogurt.
  • the invention also provides a composition comprising yogurt and a microorganism wherein the microorganism comprises a gene encoding a polypeptide according to the present invention.
  • fixation refers to the chemical conversion of carbohydrates or polypeptides in food products through the use of microorganisms. In this process carbohydrates are often convened to lactic acid.
  • sold food product refers to an edible food product prepared by a process that includes fermentation by a microorganism.
  • yogurt refers to a dairy product produced by the lactic acid fermentation of milk by the action of microorganisms.
  • recombinantly produced polypeptides refers to a polypeptide produced using recombinant DNA techniques.
  • Recombinant DNA techniques are well known and are characterized by the joining of at least two segments of DNA that are not naturally joined in nature (e.g., a bacterial promoter and a polypeptide coding sequence).
  • the reference sequence may be shorter than the full-length naturally occurring polypeptide or polynucleotide sequence but will be at least 12 residues long for the case of a polypeptide and at least 36 bases long for the case of a polynucleotide.
  • the present invention also provides methods for preparing a frozen fermented food product by adding a microorganism that is capable of fermenting the food product to produce the fermented food product and also is able to secrete the polypeptide according to the present invention.
  • the use of a microorganism that both secretes the polypeptide according to the present invention and ferments the food product has several advantages over other methods for affecting ice crystal formation and freezing temperature.
  • the claimed method avoids the costly necessity for purifying the polypeptide according to the present invention prior to addition to a food product. In addition, this will eliminate any possible contamination from the purification protocol and the pyrogenicity associated with foreign microorganisms.
  • the polypeptide according to the present invention is secret by the fermenting microorganism of the claimed invention, this process requires fewer steps than other methods.
  • the food product of the invention is usually milk but other foods that are fermented to produce an edible fermented food may also be used. Examples include cabbage (which can be fermented to produce sauerkraut), cucumbers (which can be fermented to produce pickles) and soybeans (which can be fermented to produce miso and other products).
  • the food product is contacted or mixed with a microorganism capable of fermenting the food product.
  • a microorganism capable of fermenting the food product examples include van de Guchte, 1992, FEMS Microbiology Reviews, 88:73-92).
  • the food product is milk (e.g., from a cow [i.e. bovine], ewe, mare, or goat).
  • milk e.g., from a cow [i.e. bovine], ewe, mare, or goat.
  • the action of fermenting microorganisms, typically bacteria, on the milk produces yogurt, buttermilk, or certain cheeses, according to the choice of the bacteria and the conditions of incubation.
  • the method of the invention will be used to produce yogurt from milk.
  • Yogurt is referred to by a variety of names around the world. The names and country of origin of the common varieties of yogurt are listed below:
  • yogurt is produced from either whole or skim milk from cows.
  • the milk is standardized to 10.5 to 1 1 .5% solids, heated to above 90°C. (30 to 60 minutes) to destroy any contaminating microorganisms, and then cooled.
  • the material is then inoculated with a mixed culture of Streptococcus thermophilus and Lactobacillus bulgaricus in a 1 :1 ratio. The combined action of these two organisms is usually needed to obtain the desired flavor and acid in the products.
  • Yeasts (Torulopsis holmil; Saccharomyces fragilis, cerevisiae, lactis; Candida pseudotropicalis, etc.) Fungi (Geotrichum candidum) Acetic acid bacteria (Acetobacter aceti, rasens) a Now Lactococcus lactis subsp. cremoris, Lac, lactis subsp. lactis and S. thermophilus. b Now L. mesenteroides subsp. cremoris and L. mesenteroides subsp. mesenteroides. c Now L. acidophilus, L. delbrueckii subsp. bulgaricus, L. casei subsp. casei, L.helveticus biovar. jugurti and L. delbrueckii subsp. lactis.
  • microorganisms may be genetically engineered (i.e., employing the techniques of recombinant DNA technology) so that they are able to secrete the polypeptide according to the present invention.
  • microorganisms capable of expressing and secreting an AFP can be carried out in a variety of ways that will be apparent to one of ordinary skill.
  • the DNA sequence encoding the AFP will preferably be operably linked (i.e., positioned to ensure the functioning of) to an operon which allows the DNA to be transcribed (into an RNA transcript) and translated into a polypeptide in the microorganism. Promoters for both bacteria and fungi are well known in the art.
  • Preferred operons for expression in lactic acid bacteria include the lactose operon of S. thermophilus or lac ABCDFEGX operon of L.
  • polypeptide according to the present invention may be expressed as a fusion polypeptide for increased stability or other beneficial properties.
  • polypeptide according to the present invention may be modified via a modification of the gene encoding the polypeptide.
  • modifications of the genes may be readily accomplished by a variety of well-known techniques, such as site-directed mutagenesis (see, e.g., Gillman and Smith, 1979, Gene 8:81 -97 and Roberts et al., 1987, Nature 328:731 -734).
  • the microorganisms of the invention are capable of secreting the polypeptide according to the present invention.
  • the polypeptide according to the present invention will preferably be linked to a signal peptide sequence.
  • suitable signal peptide sequences include those from the usp45 gene of L. lactis ssp lactis MG 1363 and the L. lactis ssp cremoris SK1 1 cell envelop associated protease gene (van Asseldonk et al., 1990, Gene 95:155-160; De vos et al., 1989, J. Dairy Sci. 72:3398-3405).
  • the usp45 signal peptide is preferred since it derives from the same host.
  • the polypeptide gene according to the present invention is linked to a transcription termination sequence to ensure correct termination of transcription of the polypeptide according to the present invention in the host system.
  • a gene construct including elements described above is constructed using plasmids such as pUC19, pNZ18 and pDBN183 as vectors (Solaiman et al., 1992, Plasmid, 28:25-36).
  • the gene construct is incorporated into the genome of a lactic acid bacterial species using homologous recombination techniques (Mollet et al., 1993, J. Bact., 175:4315-4324).
  • the lactic acid bacteria and E. coli strains can be maintained as recommended by Maniatis et al. in Molecular Cloning, A Laboratory Manual, supra; and Chagnand et al., 1992, Can. J. Microbiol. 38:67-74.
  • Microorganisms comprising the polypeptide according to the present invention may be applied to food products in any conventional way.
  • the bacteria or fungus can be mixed intimately with the food product that is to be fermented and frozen. It will be known by those of skill that mixtures of different microorganisms are sometimes used to produce the desired product. For example, in preparation of yogurt, S. thermophilus and L. bulgaricus are often used together.
  • the number of FAE microorganisms added to the food product will depend on the properties of the microorganisms and of the food. Generally, lactic acid FAE starter bacteria (10 10 -10 11 per ml) are incubated at 1 -5% into pasteurized and cooled milk such that the proportion results in an appropriate amount of polypeptide according to the present invention in the product.
  • the amount of AFP in the product should be an amount effective at preventing or inhibiting ice recrystallization (1 -100 mg/liter milk). This can be determined using the splat-cooling assay described by Knight et al. (1988) Cryobiology, vol. 25, pp. 55-60.
  • the fermented food product is frozen using conventional freezer operations, such as blast freezers (-20 to 40°C) or contact plate freezers (-300 to 40°C) or vacuum freeze driers. It will be apparent to one of ordinary skill that numerous variations of the aforementioned embodiments are possible.
  • Ice cream comprising the polypeptide according to the present invention
  • the present invention provides an ice cream product comprising a polypeptide according to the present invention.
  • the ice cream product can also comprise an emulsifier system together with a polypeptide according to the present invention.
  • This emulsifier system may be any system known by the skilled person.
  • the polypeptide according to the present invention may be added as a purified polypeptide, mixed with other ingredients during manufacture of the ice cream or the polypeptide according to the present invention may be present as a result of secretion from the microorganism used for fermenting the milk.
  • One way of manufacturing the ice cream according to the present invention is as follows.
  • a food intermediate is contacted with the above mentioned emulsifier system.
  • food intermediate a mixture of ingredients suitable for preparing the ice cream.
  • Ingredients suitable for preparing ice cream may include water, fat such as milkfat or vegetable fat, milk solids not fat (MSNF), sweeteners, stabilisers, flavourings and colurings.
  • MSNF milk solids not fat
  • the food intermediate comprises fat.
  • the fat is a high lauric fat or milk fat.
  • high lauric fat is meant a fat in which the predominant fatty acid is lauric acid.
  • High lauric fats such as hardened palm kernel oil and hardened coconut oil, are ⁇ ' stable.
  • the food intermediate comprises ⁇ ' stable fats.
  • the selected ingredients are mixed together.
  • the liquid ingredients are mixed together first and the dry ingredients are added subsequently.
  • the liquid ingredients may be cold or may be heated to approximately 60°C. Blending requires rapid agitation to incorporate powders and often high speed blenders are used.
  • butter/butter oil or vegetable fat is used, it should ideally be melted separately and added to the mix at 40°C or via a static mixer at the entrance of the homogeniser by means of a dosing pump.
  • Pasteurisation is carried out to destroy pathogenic bacteria and spoilage organisms such as psychrotrophs. There are three distinct stages in pasteurization: pasteurization, homogenisation and cooling
  • Homogenisation of the mix is carried out in order to form the fat emulsion by breaking down or reducing the size of the fat globules found to less than 1 ⁇ m.
  • Pasteurisation may be carried out by continuous pasteurisation or batch pasteurisation.
  • Batch pasteurisation is the old method where all mix ingredients are slowly heated in a vat equipped with a hot water jacket. In order to avoid fouling on the bottom and sides of the vat, the heating process has to be gentle with a low differential temperature (delta T) between the mix and the heating medium. As the delta T has to be low and the ratio of mix volume/vat surface is typically high, it will inevitably take several minutes just to heat the mix to a temperature of 60°C. Effective agitation of the mix is needed in order to improve the transfer of heat from the vat surface to the mix. Energy consumption for batch pasteurisation is very high and, unlike continuous pasteurisation, there is no heat recovery.
  • the mix is homogenised by means of high pressures. Homogenisation typically takes place at a temperature of about 80°C. and the homogenisation pressure can be in the region of 90 bar (1300 psi) to 250 bar (3600 psi) at a temperature of 65-75°C. Batch tanks are usually operated in tandem so that one is holding while the other is being prepared. Automatic timers and valves ensure the proper holding time has been met.
  • Homogenisation can be carried out either before or after pasteurisation.
  • the mix is cooled to refrigerated temperatures (4°C) by passing it across a heat exchanger (plate or double or triple tube).
  • the mixture is cooled to the aging temperature which is about 4°C.
  • the mix is then aged for a minimum of four hours but preferably overnight. This allows time for the fat to crystallize and for the polypeptides and polysaccharides to fully hydrate.
  • the mix may be drawn into a flavour tank where any liquid flavours, fruit purees, or colours are added.
  • the mix then enters the dynamic freezing process which both freezes a portion of the water and whips air into the frozen mix. Freezing may be carried out by a continuous freezing process or by batch freezing/whipping. Continuous freezing may be carried out in a barrel freezer.
  • the barrel freezer is a scraped-surface, tubular heat exchanger, which is jacketed with a boiling refrigerant such as ammonia or freon.
  • the mix is pumped through the barrel freezer and is drawn off the other end in about 30 seconds to 3 minutes. In the case of batch freezers the process takes 10 to 15 minutes. When the mix is drawn off the other end about 50% of its water is frozen. There are rotating blades inside the barrel freezer that keep the ice scraped off the surface of the freezer. There are also dashers inside the machine which help to whip the mix and incorporate air.
  • Ice cream contains a considerable quantity of air, typically up to half of its volume. This gives the product its characteristic lightness. The air content is termed its overrun.
  • particulate matter such as fruit pieces, nuts or cookies, may be added to the semi-frozen slurry.
  • the ice cream is then packaged and is placed into a blast freezer at -30° to -40°C where most of the remainder of the water is frozen.
  • Hardening involves static (still, quiescent) freezing of the packaged products in blast freezers.
  • the freezing rate should ideally be rapid, so freezing techniques involve low temperature (-40°C) with either enhanced convection (freezing tunnels with forced air fans) or enhanced conduction (plate freezers).
  • the ice cream may be pumped from the ice cream freezer into a low temperature extruder (single or double screw extruder) which brings the temperature of the ice cream down to -12°C to -18°C. After filling or extrusion the ice cream may be taken directly into cold storage.
  • a low temperature extruder single or double screw extruder
  • the hardened ice cream should be stored below -25°C. Below about -25°C, ice cream is quite stable for long time without danger of fast ice crystal growth.
  • the process of the present invention comprises the step of contacting a food intermediate with an emulsifier system.
  • the process comprises the step of dissolving the emulsifier system in water.
  • the emulsifier system may be dissolved in water and the food intermediate may then be contacted with water.
  • the process comprises the step of dissolving the emulsifier system in fat.
  • the emulsifier system may be dissolved in fat and the food intermediate may then be contacted with fat.
  • the process comprises a dynamic freezing step.
  • dynamic freezing step means subjecting the food intermediate to freezing conditions whilst agitating the food intermediate. This is in contrast to a quiescent freezing step in which the food intermediate is subjected to freezing conditions whilst static.
  • the process comprises a freezing step.
  • the process comprises a freezing step with a drawing temperature from the freezer lower than -4°C.
  • the drawing temperature from the freezer is about -4°C to -7°C, preferably about -5°C. to -7°C, more preferably about -5°C to -6°C, more preferably about -6°C.
  • the drawing temperature is the temperature of the ice cream as it exits the ice cream freezer.
  • Aerated food products comprising the polypeptide according to the invention
  • the present invention also provides an aerated food product comprising a polypeptide according to the invention.
  • Ice cream, sherbet, sorbet and frozen yoghurt can be mentioned as examples of food products, which may be characterized as aerated products.
  • the term "aerated” means that gas has been intentionally incorporated into a mix, for example by mechanical means.
  • the gas can be any gas, but is preferably, in the context of food products, a food-grade gas such as air, nitrogen, nitrous oxide, or carbon dioxide.
  • the aerated food products of the invention comprise a population of gas bubbles, wherein at least 65% of the gas bubbles have a diameter of less than 20 ⁇ m. Preferably at least 75%, more preferably at least 80% of the gas bubbles have a diameter of less than 20 ⁇ m. Preferably at least 50%, more preferably at least 60%, most preferably at least 75% of the gas bubbles have a diameter of less than 10 ⁇ m.
  • % overrun is defined in terms of the volume of the aerated product and the volume of the unaerated mix from which it was formed:
  • Overrun is measured at atmospheric pressure. The amount of overrun present in the product will vary depending on the desired product characteristics
  • the food product has an overrun of at least 20%, more preferably at least 50%, most preferably at least 80%.
  • the food product has an overrun of at most 400%, more preferably at most 200%, most preferably at most 120%.
  • the aerated food product may be manufactured by use of any process known in the art, such as for example by use of the pre-aeration route, which is a process for producing aerated food products comprising a large proportion of small gas bubbles starting from an unaerated mix.
  • a mix i.e. an aqueous solution and/or suspension
  • the aeration step must be of a sufficiently high "intensity" so that a large number of very small gas bubbles (less than 20 ⁇ m in diameter) are created.
  • the intensity of the aeration process depends on a number of factors, the most important of which are the rate of energy dissipation in the aeration step, the nature of the flow experienced by the mix and the gas bubbles in the aeration step, and the viscosity and temperature of the mix.
  • the aeration step should be long enough to achieve the desired degree of aeration (i.e. overrun).
  • the effectiveness of the aeration step also depends on the nature of the flow in the aerating device. Aeration is normally achieved by initially incorporating relatively large gas bubbles which are subsequently broken up into smaller ones. Elongational flow or extensional flow is known to be particularly effective at breaking up large gas bubbles, compared to simple shear flow (see e.g. Rallinson, J. M. Ann. Rev. Fluid Mech. 16, pp 45-66, 1984). Suitable high shear aeration processes and devices that can provide at least a component of elongational flow include: continuous whipping in a rotor-stator device such as an Oakes mixer (ET.
  • ET Oakes mixer
  • the effectiveness of the aeration step also depends on the viscosity and/or the temperature of the mix. By increasing the viscosity and/or lowering the temperature of the mix, the size reducing effect of the aeration device on the gas bubbles is increased. Furthermore, the effectiveness of the aeration step also depends on the formulation of the mix.
  • the effectiveness of the aeration step depends on the specific details of the process and apparatus used and the mix being aerated, it is within the compass of one skilled in the art to determine the appropriate process conditions in any particular situation, by considering the factors described above.
  • the proportion of very small gas bubbles can be increased by increasing the energy dissipated and/or by increasing the elongational flow component and/or by increasing the viscosity of the mix and/or by lowering the temperature of the mix.
  • the aerated mixture may optionally be subjected to freezing during and/or after aeration, for example if the final product is to be a frozen aerated product such as an ice cream or a sorbet. Freezing may take place simultaneously with aeration, for example in a scraped surface heat exchanger. Simultaneous freezing and aeration can aid the formation of small gas bubbles because of the increase in the mix viscosity as ice forms. When freezing takes place after aeration, it is preferably carried out so that little or no further gas is incorporated.
  • the ice content may be increased further by subsequent freezing operations, such as low-temperature extrusion, placing the aerated mixture in a mould immersed in a bath of cold liquid such as brine or glycol, dropping portions of the aerated mixture directly into a bath of cryogenic fluid such as liquid nitrogen or placing a container comprising the aerated mixture into a cold environment such as a freezer, blast freezer or cold store.
  • the subsequent freezing step is preferably carried out at low or zero shear so that little or no further gas is incorporated.
  • the aerated food products of the invention may comprise other ingredients conventionally found in food products, such as fats, milk or cream; oil or fat, notably in the form of an emulsified phase; sugars, salt, fruit and/or vegetable material, extract, juice, thickeners, such as polysaccharides, stabilisers, colours, flavours, chemical emulsifiers, such as monoglycerides; acids and preservatives.
  • Preferred food products include ice cream, sorbet, mousse, whipped cream, aerated beverages such as milk shakes and smoothies, low-fat spreads (e.g. having a fat content of 0-60 wt %), dressings and sauces.
  • the food product is a frozen or chilled aerated confection such as ice cream, sorbet or mousse.
  • Frozen aerated confections of the invention comprise the polypeptide according to the present invention and optionally one or more anti-freeze polypeptides other than the polypeptide according to the invention.
  • the amount of the total of anti-freeze polypeptides is typically at least about 0.0001 wt %, more preferably at least 0.0005 wt %, most preferably at least 0.001 wt %.
  • Anti-freeze polypeptides can be used at very low concentrations and therefore preferably the confections comprise less than 0.05 wt % Anti-freeze polypeptides. A preferred range is from about 0.001 to 0.01 wt %.
  • Anti freeze polypeptides can be used individually or in combination with other anti freeze polypeptides known in the area.
  • Frozen aerated products may optionally comprise coatings, such as a layer of chocolate or couverture and/or inclusions, such as nuts, fruit, toffee or chocolate pieces.
  • the fat content of the frozen aerated confection does not include fat present in the coating or inclusion.

Landscapes

  • Life Sciences & Earth Sciences (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Zoology (AREA)
  • General Health & Medical Sciences (AREA)
  • Organic Chemistry (AREA)
  • Genetics & Genomics (AREA)
  • Wood Science & Technology (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • General Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Food Science & Technology (AREA)
  • Polymers & Plastics (AREA)
  • Biomedical Technology (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Microbiology (AREA)
  • Medicinal Chemistry (AREA)
  • Biophysics (AREA)
  • Biotechnology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Environmental Sciences (AREA)
  • Dentistry (AREA)
  • Toxicology (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Animal Behavior & Ethology (AREA)
  • Nutrition Science (AREA)
  • Plant Pathology (AREA)
  • Tropical Medicine & Parasitology (AREA)
  • Insects & Arthropods (AREA)
  • Inorganic Chemistry (AREA)
  • Birds (AREA)
  • Epidemiology (AREA)
  • Dermatology (AREA)
  • Physics & Mathematics (AREA)
  • Agronomy & Crop Science (AREA)

Abstract

La présente invention porte sur de nouveaux polypeptides comprenant une aptitude de liaison à la glace conduisant à une activité de réduction ou d'inhibition de la formation et/ou de la croissance de cristaux de glace. La présente invention porte également sur un produit comestible et sur un support solide comprenant le nouveau polypeptide. De plus, la présente invention porte sur un procédé de fabrication du nouveau polypeptide et sur différentes utilisations du nouveau polypeptide.
EP08851310.6A 2007-11-21 2008-11-21 Polypeptides comprenant une activité de liaison à la glace Not-in-force EP2225267B1 (fr)

Priority Applications (2)

Application Number Priority Date Filing Date Title
DK12192989.7T DK2602263T3 (da) 2007-11-21 2008-11-21 Polypeptider omfattende en isbindende aktivitet
EP12192989.7A EP2602263B1 (fr) 2007-11-21 2008-11-21 Polypeptides comprenant une activité de liaison à la glace

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US397907P 2007-11-21 2007-11-21
DKPA200701656 2007-11-21
PCT/DK2008/050278 WO2009065415A1 (fr) 2007-11-21 2008-11-21 Polypeptides comprenant une activité de liaison à la glace

Related Child Applications (2)

Application Number Title Priority Date Filing Date
EP12192989.7A Division-Into EP2602263B1 (fr) 2007-11-21 2008-11-21 Polypeptides comprenant une activité de liaison à la glace
EP12192989.7A Division EP2602263B1 (fr) 2007-11-21 2008-11-21 Polypeptides comprenant une activité de liaison à la glace

Publications (2)

Publication Number Publication Date
EP2225267A1 true EP2225267A1 (fr) 2010-09-08
EP2225267B1 EP2225267B1 (fr) 2015-01-14

Family

ID=40445164

Family Applications (2)

Application Number Title Priority Date Filing Date
EP08851310.6A Not-in-force EP2225267B1 (fr) 2007-11-21 2008-11-21 Polypeptides comprenant une activité de liaison à la glace
EP12192989.7A Not-in-force EP2602263B1 (fr) 2007-11-21 2008-11-21 Polypeptides comprenant une activité de liaison à la glace

Family Applications After (1)

Application Number Title Priority Date Filing Date
EP12192989.7A Not-in-force EP2602263B1 (fr) 2007-11-21 2008-11-21 Polypeptides comprenant une activité de liaison à la glace

Country Status (9)

Country Link
US (3) US8859230B2 (fr)
EP (2) EP2225267B1 (fr)
JP (2) JP5474809B2 (fr)
KR (1) KR101700711B1 (fr)
CN (2) CN101918437B (fr)
AU (1) AU2008328371B2 (fr)
CA (1) CA2706550C (fr)
DK (2) DK2225267T3 (fr)
WO (1) WO2009065415A1 (fr)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112715897A (zh) * 2019-10-14 2021-04-30 江西巨荣生物科技有限公司 一种固化酶蜂蜜及其制备方法

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DK2225267T3 (en) 2007-11-21 2015-04-27 Roskilde Uni Polypeptides comprising a isbindende activity
GB0808350D0 (en) * 2008-05-09 2008-06-18 Airbus Uk Ltd Self-cleaning surfaces
US10039777B2 (en) 2012-03-20 2018-08-07 Neuro-Lm Sas Methods and pharmaceutical compositions of the treatment of autistic syndrome disorders
US10426159B1 (en) * 2012-05-24 2019-10-01 The Trustees Of California State University Highly efficient enhancers for antifreeze protein activity
KR101492434B1 (ko) * 2012-08-02 2015-02-23 한국해양과학기술원 플라보박테리움 프리고리스 유래 결빙방지 단백질
US20140050836A1 (en) * 2012-08-14 2014-02-20 Conopco, Inc., D/B/A Unilever Stabilized Aerated Frozen Confection Containing Hydrophobin
WO2014202089A2 (fr) * 2013-06-18 2014-12-24 Roskilde Universitet Variants de polypeptides antigel
CN105530953A (zh) * 2013-10-03 2016-04-27 日东电工株式会社 注射疫苗组合物
CA2923026A1 (fr) * 2013-10-03 2015-04-09 Nitto Denko Corporation Composition de vaccin muqueux nasal
KR20160058772A (ko) 2013-10-03 2016-05-25 닛토덴코 가부시키가이샤 점막 백신 조성물
MX2016003321A (es) * 2013-10-03 2016-08-12 Nitto Denko Corp Composicion de vacuna para mucosa.
MX370305B (es) 2013-12-02 2019-12-09 Dsm Ip Assets Bv Proteína estructurante de hielo.
CN103898013B (zh) * 2014-03-14 2015-12-02 福建农林大学 一株海旋菌及κ-卡拉胶酶的制备
US10101265B1 (en) 2014-11-07 2018-10-16 Board Of Regents For The University Of Nebraska Birefringence imaging chromatography based on highly ordered 3D nanostructures
US10519521B2 (en) * 2015-03-12 2019-12-31 The Trustees Of California State University Method of preventing carbohydrate crystallization
US20180064124A1 (en) * 2015-03-20 2018-03-08 Optifreeze Ab Apparatus and method for extending shelf life of a food product comprising water and soft tissue
EP3284795B1 (fr) * 2015-04-16 2023-08-02 The School Corporation Kansai University Utilisation d'un polymère contenant une base purique comme activateur anti-glaciogène
US20180160695A1 (en) 2015-06-02 2018-06-14 Dsm Ip Assets B.V. Use of ice structuring protein afp19 expressed in filamentous fungal strains for preparing food
CN106544385B (zh) * 2015-09-16 2020-11-27 上海理工大学 胶原抗冻肽的分离方法
CN105454222B (zh) * 2016-02-02 2018-10-30 河南省银丰生物工程技术有限公司 一种细胞冻存液及其制备方法和应用
KR102332124B1 (ko) * 2017-03-23 2021-11-26 부경대학교 산학협력단 결빙 제어용 펩타이드 표면 개질된 나노입자
EP3638049B1 (fr) * 2017-06-15 2020-12-30 Unilever PLC Emballage de produit de confiserie congelé
WO2019147674A1 (fr) * 2018-01-24 2019-08-01 Microtek Laboratories, Inc. Gels de refroidissement thermique à base d'eau comprenant un modificateur de viscosité et une protéine de nucléation de la glace
CN108802216B (zh) * 2018-05-23 2021-04-16 山东出入境检验检疫局检验检疫技术中心 一种使用区域敏感性蛋白组和/或多肽组鉴别刺参产地的方法
CN108990964B (zh) * 2018-08-09 2022-01-28 华东理工大学 细胞冻存液
KR102361781B1 (ko) 2019-10-15 2022-02-10 고려대학교 산학협력단 펩티드가 결합된 금 나노입자를 포함하는 항-동결 조성물
CN111066776A (zh) * 2019-11-09 2020-04-28 无锡市人民医院 一种卵子冷冻保护剂及其应用
US11800864B2 (en) * 2019-12-03 2023-10-31 Seoul National University R&Db Foundation Composition for inhibiting ice recrystallization
CN113430136B (zh) * 2021-07-09 2022-05-20 华中农业大学 菌株Rhodanobacter sp.LX-100及其在氧化有机硒或单质硒中的应用
CN114949037B (zh) * 2022-07-14 2023-04-21 北京农学院 黄芩黄酮类提取物在抑制鸭源鸡杆菌中的用途
CN116486903B (zh) * 2023-04-17 2023-12-29 深圳新锐基因科技有限公司 基于同源蛋白序列进化方向结合自由能变提高蛋白稳定性的方法及装置

Family Cites Families (78)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4331647A (en) 1980-03-03 1982-05-25 Goldenberg Milton David Tumor localization and therapy with labeled antibody fragments specific to tumor-associated markers
JPS57145101A (en) 1980-12-30 1982-09-08 Nat Res Dev Polymerization
DK368882A (da) 1981-08-25 1983-02-26 Alan John Kingsman Expessions vektorer
US4486533A (en) 1982-09-02 1984-12-04 St. Louis University Filamentous fungi functional replicating extrachromosomal element
US4599311A (en) 1982-08-13 1986-07-08 Kawasaki Glenn H Glycolytic promotersfor regulated protein expression: protease inhibitor
US4977092A (en) 1985-06-26 1990-12-11 Amgen Expression of exogenous polypeptides and polypeptide products including hepatitis B surface antigen in yeast cells
US5139936A (en) 1983-02-28 1992-08-18 Collaborative Research, Inc. Use of the GAL1 yeast promoter
US4661454A (en) 1983-02-28 1987-04-28 Collaborative Research, Inc. GAL1 yeast promoter linked to non galactokinase gene
US4870008A (en) 1983-08-12 1989-09-26 Chiron Corporation Secretory expression in eukaryotes
US4931373A (en) 1984-05-25 1990-06-05 Zymogenetics, Inc. Stable DNA constructs for expression of α-1 antitrypsin
US4766073A (en) 1985-02-25 1988-08-23 Zymogenetics Inc. Expression of biologically active PDGF analogs in eucaryotic cells
ATE54167T1 (de) 1984-12-06 1990-07-15 Fina Research Promotoren fuer die expression von fremden genen in hefe, plasmide, die diese promotoren enthalten, sowie deren verwendung zur herstellung von polypeptiden.
US5998200A (en) 1985-06-14 1999-12-07 Duke University Anti-fouling methods using enzyme coatings
US4882279A (en) 1985-10-25 1989-11-21 Phillips Petroleum Company Site selective genomic modification of yeast of the genus pichia
US4935349A (en) 1986-01-17 1990-06-19 Zymogenetics, Inc. Expression of higher eucaryotic genes in aspergillus
GB8611832D0 (en) 1986-05-15 1986-06-25 Holland I B Polypeptide
US4946778A (en) 1987-09-21 1990-08-07 Genex Corporation Single polypeptide chain binding molecules
US5063154A (en) 1987-06-24 1991-11-05 Whitehead Institute For Biomedical Research Pheromone - inducible yeast promoter
US5637677A (en) 1987-07-16 1997-06-10 The Trustees Of The University Of Pennsylvania Biologically active compounds and methods of constructing and using the same
US5037743A (en) 1988-08-05 1991-08-06 Zymogenetics, Inc. BAR1 secretion signal
US5223409A (en) 1988-09-02 1993-06-29 Protein Engineering Corp. Directed evolution of novel binding proteins
US5530101A (en) 1988-12-28 1996-06-25 Protein Design Labs, Inc. Humanized immunoglobulins
US5162228A (en) 1988-12-28 1992-11-10 Takeda Chemical Industries, Ltd. Gylceraldehyde-3-phosphate dehydrogenase gene and promoter
US5118792A (en) 1989-05-10 1992-06-02 Dna Plant Technology Corporation Ice crystal growth suppression polypeptides and method of making
WO1991010361A1 (fr) 1990-01-17 1991-07-25 The Regents Of The University Of California Composition ameliorant la survie de materiaux biologiques
US5358931A (en) * 1990-01-17 1994-10-25 The Regents Of The University Of California Interaction of thermal hysteresis proteins with cells and cell membranes and associated applications
SG46445A1 (en) 1990-01-26 1998-02-20 Immunomedics Inc Vaccines against cancer and infectious diseases
IL99552A0 (en) 1990-09-28 1992-08-18 Ixsys Inc Compositions containing procaryotic cells,a kit for the preparation of vectors useful for the coexpression of two or more dna sequences and methods for the use thereof
US5208146A (en) 1990-11-05 1993-05-04 The Regents Of The University Of California Murine monoclonal anti-idiotype antibodies
GB9110123D0 (en) 1991-05-10 1991-07-03 Dow Corning Organosilicon compounds their preparation and use
US5852172A (en) 1991-06-13 1998-12-22 University Of Waterloo Cold tolerances in plants
US5298418A (en) 1991-09-16 1994-03-29 Boyce Thompson Institute For Plant Research, Inc. Cell line isolated from larval midgut tissue of Trichoplusia ni
CA2150371C (fr) * 1992-12-11 2002-09-17 Dov Borovsky Pesticides et mode d'emploi
AU689214B2 (en) 1994-02-01 1998-03-26 United States Of America, Represented By The Secretary, Department Of Health And Human Services, The Fusion proteins that include antibody and nonantibody portions
US6117679A (en) 1994-02-17 2000-09-12 Maxygen, Inc. Methods for generating polynucleotides having desired characteristics by iterative selection and recombination
US5676985A (en) 1994-10-12 1997-10-14 Hsc Research And Development Limited Partnership Antifreeze polypeptide-expressing microorganisms useful in fermentation and freezing of foods
US5641655A (en) 1994-11-30 1997-06-24 Zymogenetics, Inc. Methods for producing thrombopoietin polypeptides using a mammalian tissue plasminogen activator secretory peptide
JP3739794B2 (ja) 1995-03-29 2006-01-25 カート ディー. ジョーンズ 液体供給用点滴器アセンブリ及び冷凍室への液体組成物供給方法
SK179497A3 (en) * 1995-07-05 1998-07-08 Unilever Nv Method for producing recombinant polypeptides or proteins and a food product containing them
US5716808A (en) 1995-11-09 1998-02-10 Zymogenetics, Inc. Genetic engineering of pichia methanolica
WO1997017450A2 (fr) 1995-11-09 1997-05-15 Zymogenetics, Inc. Compositions et procedes de production de polypeptides heterologues sur pichia methanolica
US5868886A (en) * 1995-12-22 1999-02-09 Alston; Mark S. Z-pin reinforced bonded composite repairs
US5723125A (en) 1995-12-28 1998-03-03 Tanox Biosystems, Inc. Hybrid with interferon-alpha and an immunoglobulin Fc linked through a non-immunogenic peptide
EP0788745A1 (fr) 1996-02-09 1997-08-13 Societe Des Produits Nestle S.A. Agents d'inhibition de la croissance des cristaux de la glace
EP0843010A1 (fr) 1996-11-19 1998-05-20 Unilever Plc Polypeptides antigel de carotte
US5736383A (en) 1996-08-26 1998-04-07 Zymogenetics, Inc. Preparation of Pichia methanolica auxotrophic mutants
AU708572B2 (en) 1996-07-17 1999-08-05 Zymogenetics Inc. Preparation of (pichia methanolica) auxotrophic mutants
AU718510B2 (en) 1996-07-17 2000-04-13 Zymogenetics Inc. Transformation of pichia methanolica
JP4175520B2 (ja) * 1996-07-26 2008-11-05 ユニリーバー・ナームローゼ・ベンノートシャープ 冷凍菓子製品
ATE287451T1 (de) 1996-07-26 2005-02-15 Unilever Nv Gefrorenes lebensmittelprodukt das hitzestabiles gefrierschutzprotein enthält
GB2315661B (en) 1996-07-26 2000-05-03 Unilever Plc Frozen food product
HUP0001550A3 (en) 1997-03-14 2001-02-28 Unilever Nv Frozen food product
GB9801420D0 (en) 1998-01-22 1998-03-18 Unilever Plc Frozen food product
GB9801408D0 (en) 1998-01-22 1998-03-18 Unilever Plc Frozen food product
BR0008885B1 (pt) 1999-03-10 2013-02-05 confeito gelado nço-aerado.
GB9929696D0 (en) 1999-12-15 2000-02-09 Unilever Plc Processes and organisms for the production of anti-freeze proteins
KR100760270B1 (ko) 2000-03-24 2007-09-19 바이오로커스 에이/에스 로진과 효소를 포함하는 방오 도료 조성물
AU2002341541A1 (en) * 2001-06-22 2003-03-03 Syngenta Participations Ag Abiotic stress responsive polynucleotides and polypeptides
JP4228068B2 (ja) * 2001-11-21 2009-02-25 独立行政法人産業技術総合研究所 魚類由来の不凍タンパク質
EP1539206A1 (fr) 2002-09-07 2005-06-15 Beiersdorf AG Preparations cosmetiques ou dermatologiques contenant des proteines antigel et/ou des glycoproteines antigel
NZ538616A (en) 2002-09-09 2008-04-30 Genesis Res & Dev Corp Ltd Antifreeze proteins isolated from forage grasses and methods for their use
US7968767B2 (en) * 2002-09-09 2011-06-28 Jeroen Demmer Antifreeze proteins isolated from forage grasses and methods for their use
US6824776B2 (en) 2003-04-16 2004-11-30 The United States Of America As Represented By The Secretary Of The Navy Silica mesoporous aerogels having three-dimensional nanoarchitecture with colloidal gold-protein superstructures nanoglued therein
WO2009094401A2 (fr) 2008-01-22 2009-07-30 Ceres, Inc. Séquences de nucléotides, et polypeptides codés par elles modifiant les caractéristiques de la réponse au froid de plantes
US20050129810A1 (en) 2003-12-10 2005-06-16 Good Humor- Breyers Ice Cream Division Of Conopco Inc Frozen confectionery product
CN1889851A (zh) 2003-12-10 2007-01-03 荷兰联合利华有限公司 含有冰结构蛋白的冷冻糖食产品
GB0329517D0 (en) 2003-12-19 2004-01-28 Danisco Process
WO2005095586A1 (fr) 2004-03-31 2005-10-13 Shionogi & Co., Ltd. Composition pour stockage par congélation de cellules contenant un dérivé incongelable de glycoprotéine
KR101226156B1 (ko) 2004-07-16 2013-01-24 듀폰 뉴트리션 바이오사이언시즈 에이피에스 효소적 오일-탈검 방법
AU2005286427B2 (en) * 2004-09-24 2011-09-15 Basf Plant Science Gmbh Plant cells and plants with increased tolerance to environmental stress
CN101039588B (zh) 2004-10-18 2011-02-09 荷兰联合利华有限公司 低脂冷冻甜食产品
GB0508865D0 (en) 2005-04-29 2005-06-08 Danisco Product
ES2306039T3 (es) 2005-12-21 2008-11-01 Unilever N.V. Dulce aireado congelado.
ATE443451T1 (de) 2006-08-07 2009-10-15 Unilever Nv Eiskonfekt
GB0622580D0 (en) 2006-11-13 2006-12-20 Danisco Method
DE602006020943D1 (de) 2006-11-27 2011-05-05 Palsgaard As Gefrorene nahrungsmittel, emulgatorsysteme und damit in zusammenhang stehende verfahren
BRPI0705417B1 (pt) 2006-12-20 2016-08-16 Unilever Nv produto alimentício aerado e processos para a produção de um produto alimentício aerado
DK2225267T3 (en) 2007-11-21 2015-04-27 Roskilde Uni Polypeptides comprising a isbindende activity

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2009065415A1 *

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
CN112715897A (zh) * 2019-10-14 2021-04-30 江西巨荣生物科技有限公司 一种固化酶蜂蜜及其制备方法

Also Published As

Publication number Publication date
JP6043986B2 (ja) 2016-12-14
DK2225267T3 (en) 2015-04-27
US9241511B2 (en) 2016-01-26
WO2009065415A1 (fr) 2009-05-28
CN101918437B (zh) 2013-08-14
AU2008328371A1 (en) 2009-05-28
CN101918437A (zh) 2010-12-15
US10266576B2 (en) 2019-04-23
JP2014113164A (ja) 2014-06-26
KR101700711B1 (ko) 2017-01-31
CN103059118A (zh) 2013-04-24
JP2011504107A (ja) 2011-02-03
EP2602263A2 (fr) 2013-06-12
CA2706550A1 (fr) 2009-05-28
JP5474809B2 (ja) 2014-04-16
CN103059118B (zh) 2017-11-24
US20160215029A1 (en) 2016-07-28
KR20100110778A (ko) 2010-10-13
EP2225267B1 (fr) 2015-01-14
US20100312045A1 (en) 2010-12-09
EP2602263A3 (fr) 2013-11-13
CA2706550C (fr) 2021-12-14
EP2602263B1 (fr) 2019-07-17
US20150079254A1 (en) 2015-03-19
US8859230B2 (en) 2014-10-14
DK2602263T3 (da) 2019-10-21
AU2008328371B2 (en) 2011-08-11

Similar Documents

Publication Publication Date Title
US10266576B2 (en) Polypeptides comprising an ice-binding activity
Griffith et al. Antifreeze proteins and their potential use in frozen foods
Venketesh et al. Properties, potentials, and prospects of antifreeze proteins
WO2014202089A2 (fr) Variants de polypeptides antigel
US6348569B1 (en) Spruce budworm antifreeze proteins, genes and method of using same
CA2296005C (fr) Proteines a hysterese thermique (antigel) tirees du tenebrion meunier
Kawahara Characterizations of functions of biological materials having controlling-ability against ice crystal growth
Lillford et al. In vitro uses of biological cryoprotectants
AU2011250724B2 (en) Polypeptides comprising an ice-binding activity
JP2004344033A (ja) 魚類が有する不凍タンパク質

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20100621

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

AX Request for extension of the european patent

Extension state: AL BA MK RS

17Q First examination report despatched

Effective date: 20101116

DAX Request for extension of the european patent (deleted)
REG Reference to a national code

Ref country code: DE

Ref legal event code: R079

Ref document number: 602008036390

Country of ref document: DE

Free format text: PREVIOUS MAIN CLASS: C07K0014435000

Ipc: A01N0001020000

GRAP Despatch of communication of intention to grant a patent

Free format text: ORIGINAL CODE: EPIDOSNIGR1

RIC1 Information provided on ipc code assigned before grant

Ipc: A01N 1/02 20060101AFI20141024BHEP

Ipc: A23G 9/38 20060101ALI20141024BHEP

Ipc: C07K 14/435 20060101ALI20141024BHEP

GRAS Grant fee paid

Free format text: ORIGINAL CODE: EPIDOSNIGR3

INTG Intention to grant announced

Effective date: 20141110

GRAA (expected) grant

Free format text: ORIGINAL CODE: 0009210

AK Designated contracting states

Kind code of ref document: B1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MT NL NO PL PT RO SE SI SK TR

REG Reference to a national code

Ref country code: GB

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: CH

Ref legal event code: EP

REG Reference to a national code

Ref country code: IE

Ref legal event code: FG4D

REG Reference to a national code

Ref country code: AT

Ref legal event code: REF

Ref document number: 706513

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150215

REG Reference to a national code

Ref country code: DE

Ref legal event code: R096

Ref document number: 602008036390

Country of ref document: DE

Effective date: 20150305

REG Reference to a national code

Ref country code: DK

Ref legal event code: T3

Effective date: 20150420

REG Reference to a national code

Ref country code: SE

Ref legal event code: TRGR

REG Reference to a national code

Ref country code: NL

Ref legal event code: VDEP

Effective date: 20150114

REG Reference to a national code

Ref country code: AT

Ref legal event code: MK05

Ref document number: 706513

Country of ref document: AT

Kind code of ref document: T

Effective date: 20150114

REG Reference to a national code

Ref country code: LT

Ref legal event code: MG4D

REG Reference to a national code

Ref country code: NO

Ref legal event code: T2

Effective date: 20150114

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BG

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150414

Ref country code: FI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: HR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: LT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: ES

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: LV

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: IS

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150514

Ref country code: PL

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: AT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: GR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150415

REG Reference to a national code

Ref country code: DE

Ref legal event code: R097

Ref document number: 602008036390

Country of ref document: DE

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 8

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CZ

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: EE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: SK

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: RO

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

PLBE No opposition filed within time limit

Free format text: ORIGINAL CODE: 0009261

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: NO OPPOSITION FILED WITHIN TIME LIMIT

26N No opposition filed

Effective date: 20151015

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SI

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: BE

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: LU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20151121

Ref country code: MC

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

REG Reference to a national code

Ref country code: IE

Ref legal event code: MM4A

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20151121

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 9

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: HU

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT; INVALID AB INITIO

Effective date: 20081121

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: CY

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: MT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

Ref country code: TR

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 10

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: PT

Free format text: LAPSE BECAUSE OF FAILURE TO SUBMIT A TRANSLATION OF THE DESCRIPTION OR TO PAY THE FEE WITHIN THE PRESCRIBED TIME-LIMIT

Effective date: 20150114

REG Reference to a national code

Ref country code: FR

Ref legal event code: PLFP

Year of fee payment: 11

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: DK

Payment date: 20211118

Year of fee payment: 14

Ref country code: FR

Payment date: 20211118

Year of fee payment: 14

Ref country code: NO

Payment date: 20211117

Year of fee payment: 14

Ref country code: GB

Payment date: 20211118

Year of fee payment: 14

Ref country code: SE

Payment date: 20211117

Year of fee payment: 14

Ref country code: DE

Payment date: 20211115

Year of fee payment: 14

PGFP Annual fee paid to national office [announced via postgrant information from national office to epo]

Ref country code: IT

Payment date: 20211115

Year of fee payment: 14

Ref country code: CH

Payment date: 20211117

Year of fee payment: 14

REG Reference to a national code

Ref country code: DE

Ref legal event code: R119

Ref document number: 602008036390

Country of ref document: DE

REG Reference to a national code

Ref country code: DK

Ref legal event code: EBP

Effective date: 20221130

REG Reference to a national code

Ref country code: NO

Ref legal event code: MMEP

REG Reference to a national code

Ref country code: CH

Ref legal event code: PL

REG Reference to a national code

Ref country code: SE

Ref legal event code: EUG

GBPC Gb: european patent ceased through non-payment of renewal fee

Effective date: 20221121

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: NO

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

Ref country code: LI

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

Ref country code: CH

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: SE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221122

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: IT

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221121

Ref country code: GB

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221121

Ref country code: DK

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130

Ref country code: DE

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20230601

PG25 Lapsed in a contracting state [announced via postgrant information from national office to epo]

Ref country code: FR

Free format text: LAPSE BECAUSE OF NON-PAYMENT OF DUE FEES

Effective date: 20221130